Publications of Ahmad Hariri

%%    
@article{fds372228,
   Author = {Lay-Yee, R and Hariri, AR and Knodt, AR and Barrett-Young, A and Matthews, T and Milne, BJ},
   Title = {Social isolation from childhood to mid-adulthood: is there
             an association with older brain age?},
   Journal = {Psychological medicine},
   Volume = {53},
   Number = {16},
   Pages = {7874-7882},
   Year = {2023},
   Month = {December},
   Abstract = {<h4>Background</h4>Older brain age - as estimated from
             structural MRI data - is known to be associated with
             detrimental mental and physical health outcomes in older
             adults. Social isolation, which has similar detrimental
             effects on health, may be associated with accelerated brain
             aging though little is known about how different
             trajectories of social isolation across the life course
             moderate this association. We examined the associations
             between social isolation trajectories from age 5 to age 38
             and brain age assessed at age 45.<h4>Methods</h4>We
             previously created a typology of social isolation based on
             onset during the life course and persistence into adulthood,
             using group-based trajectory analysis of longitudinal data
             from a New Zealand birth cohort. The typology comprises four
             groups: 'never-isolated', 'adult-only', 'child-only', and
             persistent 'child-adult' isolation. A brain age gap estimate
             (brainAGE) - the difference between predicted age from
             structural MRI date and chronological age - was derived at
             age 45. We undertook analyses of brainAGE with trajectory
             group as the predictor, adjusting for sex, family
             socio-economic status, and a range of familial and
             child-behavioral factors.<h4>Results</h4>Older brain age in
             mid-adulthood was associated with trajectories of social
             isolation after adjustment for family and child confounders,
             particularly for the 'adult-only' group compared to the
             'never-isolated' group.<h4>Conclusions</h4>Although our
             findings are associational, they indicate that preventing
             social isolation, particularly in mid-adulthood, may help to
             avert accelerated brain aging associated with negative
             health outcomes later in life.},
   Doi = {10.1017/s0033291723001964},
   Key = {fds372228}
}

@article{fds373508,
   Author = {Knodt, AR and Elliott, ML and Whitman, ET and Winn, A and Addae, A and Ireland, D and Poulton, R and Ramrakha, S and Caspi, A and Moffitt, TE and Hariri, AR},
   Title = {Test-retest reliability and predictive utility of a
             macroscale principal functional connectivity
             gradient.},
   Journal = {Human brain mapping},
   Volume = {44},
   Number = {18},
   Pages = {6399-6417},
   Year = {2023},
   Month = {December},
   Abstract = {Mapping individual differences in brain function has been
             hampered by poor reliability as well as limited
             interpretability. Leveraging patterns of brain-wide
             functional connectivity (FC) offers some promise in this
             endeavor. In particular, a macroscale principal FC gradient
             that recapitulates a hierarchical organization spanning
             molecular, cellular, and circuit level features along a
             sensory-to-association cortical axis has emerged as both a
             parsimonious and interpretable measure of individual
             differences in behavior. However, the measurement
             reliabilities of this FC gradient have not been fully
             evaluated. Here, we assess the reliabilities of both global
             and regional principal FC gradient measures using
             test-retest data from the young adult Human Connectome
             Project (HCP-YA) and the Dunedin Study. Analyses revealed
             that the reliabilities of principal FC gradient measures
             were (1) consistently higher than those for traditional
             edge-wise FC measures, (2) higher for FC measures derived
             from general FC (GFC) in comparison with resting-state FC,
             and (3) higher for longer scan lengths. We additionally
             examined the relative utility of these principal FC gradient
             measures in predicting cognition and aging in both datasets
             as well as the HCP-aging dataset. These analyses revealed
             that regional FC gradient measures and global gradient range
             were significantly associated with aging in all three
             datasets, and moderately associated with cognition in the
             HCP-YA and Dunedin Study datasets, reflecting contractions
             and expansions of the cortical hierarchy, respectively.
             Collectively, these results demonstrate that measures of the
             principal FC gradient, especially derived using GFC,
             effectively capture a reliable feature of the human brain
             subject to interpretable and biologically meaningful
             individual variation, offering some advantages over
             traditional edge-wise FC measures in the search for
             brain-behavior associations.},
   Doi = {10.1002/hbm.26517},
   Key = {fds373508}
}

@article{fds370718,
   Author = {Strauman, TJ and Hariri, AR},
   Title = {Revising a Self-Regulation Phenotype for Depression Through
             Individual Differences in Macroscale Brain
             Organization.},
   Journal = {Current directions in psychological science},
   Volume = {32},
   Number = {4},
   Pages = {267-275},
   Year = {2023},
   Month = {August},
   Abstract = {<i>Self-regulation</i> denotes the processes by which people
             initiate, maintain, and control their own thoughts,
             behaviors, or emotions to produce a desired outcome or avoid
             an undesired outcome. Self-regulation brings the influence
             of distal factors such as biology, temperament, and
             socialization history onto cognition, motivation, and
             behavior. Dysfunction in self-regulation represents a
             contributory causal factor for psychopathology. Accordingly,
             we previously proposed a risk phenotype model for depression
             drawing from regulatory focus theory and traditional
             task-based fMRI studies. In this article, we revise and
             expand our risk phenotype model using insights from new
             methodologies allowing quantification of individual
             differences in task-free macroscale brain organization. We
             offer a set of hypotheses as examples of how examination of
             intrinsic macroscale brain organization can extend and
             enrich investigations of self-regulation and depression. In
             doing so, we hope to promote a useful heuristic for model
             development and for identifying transdiagnostic risk
             phenotypes in psychopathology.},
   Doi = {10.1177/09637214221149742},
   Key = {fds370718}
}

@article{fds371656,
   Author = {Whitman, ET and Knodt, AR and Elliott, ML and Abraham, WC and Cheyne, K and Hogan, S and Ireland, D and Keenan, R and Leung, JH and Melzer, TR and Poulton, R and Purdy, SC and Ramrakha, S and Thorne, PR and Caspi, A and Moffitt, TE and Hariri, AR},
   Title = {Functional topography of the neocortex predicts covariation
             in complex cognitive and basic motor abilities.},
   Journal = {Cerebral cortex (New York, N.Y. : 1991)},
   Volume = {33},
   Number = {13},
   Pages = {8218-8231},
   Year = {2023},
   Month = {June},
   Abstract = {Although higher-order cognitive and lower-order sensorimotor
             abilities are generally regarded as distinct and studied
             separately, there is evidence that they not only covary but
             also that this covariation increases across the lifespan.
             This pattern has been leveraged in clinical settings where a
             simple assessment of sensory or motor ability (e.g. hearing,
             gait speed) can forecast age-related cognitive decline and
             risk for dementia. However, the brain mechanisms underlying
             cognitive, sensory, and motor covariation are largely
             unknown. Here, we examined whether such covariation in
             midlife reflects variability in common versus distinct
             neocortical networks using individualized maps of functional
             topography derived from BOLD fMRI data collected in 769
             45-year-old members of a population-representative cohort.
             Analyses revealed that variability in basic motor but not
             hearing ability reflected individual differences in the
             functional topography of neocortical networks typically
             supporting cognitive ability. These patterns suggest that
             covariation in motor and cognitive abilities in midlife
             reflects convergence of function in higher-order neocortical
             networks and that gait speed may not be simply a measure of
             physical function but rather an integrative index of nervous
             system health.},
   Doi = {10.1093/cercor/bhad109},
   Key = {fds371656}
}

@article{fds362550,
   Author = {Cobb, AR and Rubin, M and Stote, DL and Baldwin, BC and Lee, H-J and Hariri, AR and Telch, MJ},
   Title = {Hippocampal volume and volume asymmetry prospectively
             predict PTSD symptom emergence among Iraq-deployed
             soldiers.},
   Journal = {Psychological medicine},
   Volume = {53},
   Number = {5},
   Pages = {1906-1913},
   Year = {2023},
   Month = {April},
   Abstract = {<h4>Background</h4>Evidence suggests a link between smaller
             hippocampal volume (HV) and post-traumatic stress disorder
             (PTSD). However, there has been little prospective research
             testing this question directly and it remains unclear
             whether smaller HV confers risk or is a consequence of
             traumatization and PTSD.<h4>Methods</h4>U.S. soldiers
             (<i>N</i> = 107) completed a battery of clinical
             assessments, including structural magnetic resonance imaging
             pre-deployment. Once deployed they completed monthly
             assessments of traumatic-stressors and symptoms. We
             hypothesized that smaller HV would potentiate the effects of
             traumatic stressors on PTSD symptoms in theater. Analyses
             evaluated whether total HV, lateral (right <i>v.</i> left)
             HV, or HV asymmetry (right - left) moderated the effects of
             stressor-exposure during deployment on PTSD
             symptoms.<h4>Results</h4>Findings revealed no interaction
             between total HV and average monthly traumatic-stressors on
             PTSD symptoms <i>b</i> = -0.028, <i>p</i> = 0.681 [95%
             confidence interval (CI) -0.167 to 0.100]. However, in the
             context of greater exposure to average monthly traumatic
             stressors, greater right HV was associated with fewer PTSD
             symptoms <i>b</i> = -0.467, <i>p</i> = 0.023 (95% CI -0.786
             to -0.013), whereas greater left HV was unexpectedly
             associated with greater PTSD symptoms <i>b</i> = 0.435,
             <i>p</i> = 0.024 (95% CI 0.028-0.715).<h4>Conclusions</h4>Our
             findings highlight the importance of considering the complex
             role of HV, in particular HV asymmetry, in predicting the
             emergence of PTSD symptoms in response to war-zone
             trauma.},
   Doi = {10.1017/s0033291721003548},
   Key = {fds362550}
}

@article{fds370934,
   Author = {Barrett-Young, A and Abraham, WC and Cheung, CY and Gale, J and Hogan,
             S and Ireland, D and Keenan, R and Knodt, AR and Melzer, TR and Moffitt,
             TE and Ramrakha, S and Tham, YC and Wilson, GA and Wong, TY and Hariri, AR and Poulton, R},
   Title = {Associations Between Thinner Retinal Neuronal Layers and
             Suboptimal Brain Structural Integrity in a Middle-Aged
             Cohort.},
   Journal = {Eye and brain},
   Volume = {15},
   Pages = {25-35},
   Year = {2023},
   Month = {January},
   Abstract = {<h4>Purpose</h4>The retina has potential as a biomarker of
             brain health and Alzheimer's disease (AD) because it is the
             only part of the central nervous system which can be easily
             imaged and has advantages over brain imaging technologies.
             Few studies have compared retinal and brain measurements in
             a middle-aged sample. The objective of our study was to
             investigate whether retinal neuronal measurements were
             associated with structural brain measurements in a
             middle-aged population-based cohort.<h4>Participants and
             methods</h4>Participants were members of the Dunedin
             Multidisciplinary Health and Development Study (n=1037; a
             longitudinal cohort followed from birth and at ages 3, 5, 7,
             9, 11, 13, 15, 18, 21, 26, 32, 38, and most recently at age
             45, when 94% of the living Study members participated).
             Retinal nerve fibre layer (RNFL) and ganglion cell-inner
             plexiform layer (GC-IPL) thickness were measured by optical
             coherence tomography (OCT). Brain age gap estimate
             (brainAGE), cortical surface area, cortical thickness,
             subcortical grey matter volumes, white matter
             hyperintensities, were measured by magnetic resonance
             imaging (MRI).<h4>Results</h4>Participants with both MRI and
             OCT data were included in the analysis (RNFL n=828, female
             n=413 [49.9%], male n=415 [50.1%]; GC-IPL n=825, female
             n=413 [50.1%], male n=412 [49.9%]). Thinner retinal neuronal
             layers were associated with older brain age, smaller
             cortical surface area, thinner average cortex, smaller
             subcortical grey matter volumes, and increased volume of
             white matter hyperintensities.<h4>Conclusion</h4>These
             findings provide evidence that the retinal neuronal layers
             reflect differences in midlife structural brain integrity
             consistent with increased risk for later AD, supporting the
             proposition that the retina may be an early biomarker of
             brain health.},
   Doi = {10.2147/eb.s402510},
   Key = {fds370934}
}

@article{fds365599,
   Author = {Knodt, AR and Meier, MH and Ambler, A and Gehred, MZ and Harrington, H and Ireland, D and Poulton, R and Ramrakha, S and Caspi, A and Moffitt, TE and Hariri, AR},
   Title = {Diminished Structural Brain Integrity in Long-term Cannabis
             Users Reflects a History of Polysubstance
             Use.},
   Journal = {Biological psychiatry},
   Volume = {92},
   Number = {11},
   Pages = {861-870},
   Year = {2022},
   Month = {December},
   Abstract = {<h4>Background</h4>Cannabis legalization and use are
             outpacing our understanding of its long-term effects on
             brain and behavior, which is fundamental for effective
             policy and health practices. Existing studies are limited by
             small samples, cross-sectional measures, failure to separate
             long-term from recreational use, and inadequate control for
             other substance use. Here, we address these limitations by
             determining the structural brain integrity of long-term
             cannabis users in the Dunedin Study, a longitudinal
             investigation of a population-representative birth cohort
             followed to midlife.<h4>Methods</h4>We leveraged prospective
             measures of cannabis, alcohol, tobacco, and other illicit
             drug use in addition to structural neuroimaging in 875 study
             members at age 45 to test for differences in both global and
             regional gray and white matter integrity between long-term
             cannabis users and lifelong nonusers. We additionally tested
             for dose-response associations between continuous measures
             of cannabis use and brain structure, including careful
             adjustments for use of other substances.<h4>Results</h4>Long-term
             cannabis users had a thinner cortex, smaller subcortical
             gray matter volumes, and higher machine learning-predicted
             brain age than nonusers. However, these differences in
             structural brain integrity were explained by the propensity
             of long-term cannabis users to engage in polysubstance use,
             especially with alcohol and tobacco.<h4>Conclusions</h4>These
             findings suggest that diminished midlife structural brain
             integrity in long-term cannabis users reflects a broader
             pattern of polysubstance use, underlining the importance of
             understanding comorbid substance use in efforts to curb the
             negative effects of cannabis on brain and behavior as well
             as establish more effective policy and health
             practices.},
   Doi = {10.1016/j.biopsych.2022.06.018},
   Key = {fds365599}
}

@article{fds367351,
   Author = {Meier, MH and Caspi, A and Ambler, A and Hariri, AR and Harrington, H and Hogan, S and Houts, R and Knodt, AR and Ramrakha, S and Richmond-Rakerd,
             LS and Poulton, R and Moffitt, TE},
   Title = {Preparedness for healthy ageing and polysubstance use in
             long-term cannabis users: a population-representative
             longitudinal study.},
   Journal = {The lancet. Healthy longevity},
   Volume = {3},
   Number = {10},
   Pages = {e703-e714},
   Year = {2022},
   Month = {October},
   Abstract = {<h4>Background</h4>Cannabis is often characterised as a
             young person's drug. However, people who began consuming
             cannabis in the 1970s and 1980s are no longer young and some
             have consumed it for many years. This study tested the
             preregistered hypothesis that long-term cannabis users show
             accelerated biological ageing in midlife and poorer health
             preparedness, financial preparedness, and social
             preparedness for old age.<h4>Methods</h4>In this
             longitudinal study, participants comprised a
             population-representative cohort of 1037 individuals born in
             Dunedin, New Zealand, between April, 1972, and March, 1973,
             and followed to age 45 years. Cannabis, tobacco, and alcohol
             use and dependence were assessed at ages 18 years, 21 years,
             26 years, 32 years, 38 years, and 45 years. Biological
             ageing and health, financial, and social preparedness for
             old age were assessed at age 45 years. Long-term cannabis
             users were compared using independent samples t tests with
             five groups: lifelong cannabis non-users, long-term tobacco
             users, long-term alcohol users, midlife recreational
             cannabis users, and cannabis quitters. In addition,
             regression analyses tested dose-response associations for
             continuously measured persistence of cannabis dependence
             from age 18 years to 45 years, with associations adjusted
             for sex, childhood socioeconomic status, childhood IQ, low
             childhood self-control, family substance dependence history,
             and persistence of alcohol, tobacco, and other illicit drug
             dependence.<h4>Findings</h4>Of 997 cohort members still
             alive at age 45 years, 938 (94%) were assessed at age 45
             years. Long-term cannabis users showed statistically
             significant accelerated biological ageing and were less
             equipped to manage a range of later-life health, financial,
             and social demands than non-users. Standardised mean
             differences between long-term cannabis users and non-users
             were large: 0·70 (95% CI 0·46 to 0·94; p<0·0001) for
             biological ageing, -0·72 (-0·96 to -0·49, p<0·0001) for
             health preparedness, -1·08 (-1·31 to -0·85; p<0·0001)
             for financial preparedness, and -0·59 (-0·84 to -0·34,
             p<0·0001) for social preparedness. Long-term cannabis users
             did not fare better than long-term tobacco or alcohol users.
             Tests of dose-response associations suggested that cannabis
             associations could not be explained by the socioeconomic
             origins, childhood IQ, childhood self-control, and family
             substance-dependence history of long-term cannabis users.
             Statistical adjustment for long-term tobacco, alcohol, and
             other illicit drug dependence suggested that long-term
             cannabis users' tendency toward polysubstance dependence
             accounted for their accelerated biological ageing and poor
             financial and health preparedness, although not for their
             poor social preparedness (β -0·10, 95% CI -0·18 to
             -0·02; p=0·017).<h4>Interpretation</h4>Long-term cannabis
             users are underprepared for the demands of old age. Although
             long-term cannabis use appears detrimental, the greatest
             challenge to healthy ageing is not use of any specific
             substance, but rather the long-term polysubstance use that
             characterises many long-term cannabis users. Substance-use
             interventions should include practical strategies for
             improving health and building financial and social capital
             for healthy longevity.<h4>Funding</h4>The National Institute
             on Aging and the UK Medical Research Council. The Dunedin
             Research Unit is supported by the New Zealand Health
             Research Council and the New Zealand Ministry of Business,
             Innovation and Employment.},
   Doi = {10.1016/s2666-7568(22)00201-x},
   Key = {fds367351}
}

@article{fds366147,
   Author = {Kim, MJ and Knodt, AR and Hariri, AR},
   Title = {Meta-analytic activation maps can help identify affective
             processes captured by contrast-based task fMRI: the case of
             threat-related facial expressions.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {17},
   Number = {9},
   Pages = {777-787},
   Year = {2022},
   Month = {September},
   Abstract = {Meta-analysis of functional magnetic resonance imaging
             (fMRI) data is an effective method for capturing the
             distributed patterns of brain activity supporting discrete
             cognitive and affective processes. One opportunity presented
             by the resulting meta-analysis maps (MAMs) is as a reference
             for better understanding the nature of individual contrast
             maps (ICMs) derived from specific task fMRI data. Here, we
             compared MAMs from 148 neuroimaging studies representing
             emotion categories of fear, anger, disgust, happiness and
             sadness with ICMs from fearful > neutral and
             angry > neutral faces from an independent dataset of
             task fMRI (n = 1263). Analyses revealed that both fear
             and anger ICMs exhibited the greatest pattern similarity to
             fear MAMs. As the number of voxels included for the
             computation of pattern similarity became more selective, the
             specificity of MAM-ICM correspondence decreased. Notably,
             amygdala activity long considered critical for processing
             threat-related facial expressions was neither sufficient nor
             necessary for detecting MAM-ICM pattern similarity effects.
             Our analyses suggest that both fearful and angry facial
             expressions are best captured by distributed patterns of
             brain activity, a putative neural correlate of threat. More
             generally, our analyses demonstrate how MAMs can be
             leveraged to better understand affective processes captured
             by ICMs in task fMRI data.},
   Doi = {10.1093/scan/nsac010},
   Key = {fds366147}
}

@article{fds367352,
   Author = {Sugden, K and Caspi, A and Elliott, ML and Bourassa, KJ and Chamarti, K and Corcoran, DL and Hariri, AR and Houts, RM and Kothari, M and Kritchevsky, S and Kuchel, GA and Mill, JS and Williams, BS and Belsky,
             DW and Moffitt, TE and Alzheimer's Disease Neuroimaging
             Initiative*},
   Title = {Association of Pace of Aging Measured by Blood-Based DNA
             Methylation With Age-Related Cognitive Impairment and
             Dementia.},
   Journal = {Neurology},
   Volume = {99},
   Number = {13},
   Pages = {e1402-e1413},
   Year = {2022},
   Month = {September},
   Abstract = {<h4>Background and objectives</h4>DNA methylation algorithms
             are increasingly used to estimate biological aging; however,
             how these proposed measures of whole-organism biological
             aging relate to aging in the brain is not known. We used
             data from the Alzheimer's Disease Neuroimaging Initiative
             (ADNI) and the Framingham Heart Study (FHS) Offspring Cohort
             to test the association between blood-based DNA methylation
             measures of biological aging and cognitive impairment and
             dementia in older adults.<h4>Methods</h4>We tested 3
             "generations" of DNA methylation age algorithms (first
             generation: Horvath and Hannum clocks; second generation:
             PhenoAge and GrimAge; and third generation: DunedinPACE,
             Dunedin Pace of Aging Calculated from the Epigenome) against
             the following measures of cognitive impairment in ADNI:
             clinical diagnosis of dementia and mild cognitive
             impairment, scores on Alzheimer disease (AD) / Alzheimer
             disease and related dementias (ADRD) screening tests
             (Alzheimer's Disease Assessment Scale, Mini-Mental State
             Examination, and Montreal Cognitive Assessment), and scores
             on cognitive tests (Rey Auditory Verbal Learning Test,
             Logical Memory test, and Trail Making Test). In an
             independent replication in the FHS Offspring Cohort, we
             further tested the longitudinal association between the DNA
             methylation algorithms and the risk of developing
             dementia.<h4>Results</h4>In ADNI (<i>N</i> = 649
             individuals), the first-generation (Horvath and Hannum DNA
             methylation age clocks) and the second-generation (PhenoAge
             and GrimAge) DNA methylation measures of aging were not
             consistently associated with measures of cognitive
             impairment in older adults. By contrast, a third-generation
             measure of biological aging, DunedinPACE, was associated
             with clinical diagnosis of Alzheimer disease (beta [95% CI]
             = 0.28 [0.08-0.47]), poorer scores on Alzheimer disease/ADRD
             screening tests (beta [Robust SE] = -0.10 [0.04] to
             0.08[0.04]), and cognitive tests (beta [Robust SE] = -0.12
             [0.04] to 0.10 [0.03]). The association between faster pace
             of aging, as measured by DunedinPACE, and risk of developing
             dementia was confirmed in a longitudinal analysis of the FHS
             Offspring Cohort (<i>N</i> = 2,264 individuals, hazard ratio
             [95% CI] = 1.27 [1.07-1.49]).<h4>Discussion</h4>Third-generation
             blood-based DNA methylation measures of aging could prove
             valuable for measuring differences between individuals in
             the rate at which they age and in their risk for cognitive
             decline, and for evaluating interventions to slow
             aging.},
   Doi = {10.1212/wnl.0000000000200898},
   Key = {fds367352}
}

@article{fds364214,
   Author = {Farber, MJ and Gee, DG and Hariri, AR},
   Title = {Normative range parenting and the developing brain: A
             scoping review and recommendations for future
             research.},
   Journal = {The European journal of neuroscience},
   Volume = {55},
   Number = {9-10},
   Pages = {2341-2358},
   Year = {2022},
   Month = {May},
   Abstract = {Studies of early adversity such as trauma, abuse, and
             neglect highlight the critical importance of quality
             caregiving in brain development and mental health. However,
             the impact of normative range variability in caregiving on
             such biobehavioral processes remains poorly understood.
             Thus, we lack an essential foundation for understanding
             broader, population-representative developmental mechanisms
             of risk and resilience. Here, we conduct a scoping review of
             the extant literature centered on the question, "Is
             variability in normative range parenting associated with
             variability in brain structure and function?" After removing
             duplicates and screening by title, abstract, and full-text,
             23 records were included in a qualitative review. The most
             striking outcome of this review was not only how few studies
             have explored associations between brain development and
             normative range parenting, but also how little
             methodological consistency exists across published studies.
             In light of these limitations, we propose recommendations
             for future research on normative range parenting and brain
             development. In doing so, we hope to facilitate
             evidence-based research that will help inform policies and
             practices that yield optimal developmental trajectories and
             mental health as well as extend the literature on the
             neurodevelopmental impact of early life stress.},
   Doi = {10.1111/ejn.15003},
   Key = {fds364214}
}

@article{fds364180,
   Author = {Kim, MJ and Elliott, ML and Knodt, AR and Hariri,
             AR},
   Title = {A Connectome-wide Functional Signature of Trait
             Anger.},
   Journal = {Clinical psychological science : a journal of the
             Association for Psychological Science},
   Volume = {10},
   Number = {3},
   Pages = {584-592},
   Year = {2022},
   Month = {May},
   Abstract = {Past research on the brain correlates of trait anger has
             been limited by small sample sizes, a focus on relatively
             few regions-of-interest, and poor test-retest reliability of
             functional brain measures. To address these limitations, we
             conducted a data-driven analysis of variability in
             connectome-wide functional connectivity in a sample of 1,048
             young adult volunteers. Multi-dimensional matrix regression
             analysis showed that self-reported trait anger maps onto
             variability in the whole-brain functional connectivity
             patterns of three brain regions that serve action-related
             functions: bilateral supplementary motor area (SMA) and the
             right lateral frontal pole. We then demonstrate trait anger
             modulates the functional connectivity of these regions with
             canonical brain networks supporting somatomotor, affective,
             self-referential, and visual information processes. Our
             findings offer novel neuroimaging evidence for interpreting
             trait anger as a greater propensity to provoked action,
             supporting ongoing efforts to understand its utility as a
             potential transdiagnostic marker for disordered states
             characterized by aggressive behavior.},
   Doi = {10.1177/21677026211030240},
   Key = {fds364180}
}

@article{fds364143,
   Author = {Meier, MH and Caspi, A and R Knodt and A and Hall, W and Ambler, A and Harrington, H and Hogan, S and M Houts and R and Poulton, R and Ramrakha,
             S and Hariri, AR and Moffitt, TE},
   Title = {Long-Term Cannabis Use and Cognitive Reserves and
             Hippocampal Volume in Midlife.},
   Journal = {The American journal of psychiatry},
   Volume = {179},
   Number = {5},
   Pages = {362-374},
   Year = {2022},
   Month = {May},
   Abstract = {<h4>Objective</h4>Cannabis use is increasing among midlife
             and older adults. This study tested the hypotheses that
             long-term cannabis use is associated with cognitive deficits
             and smaller hippocampal volume in midlife, which is
             important because midlife cognitive deficits and smaller
             hippocampal volume are risk factors for dementia.<h4>Methods</h4>Participants
             are members of a representative cohort of 1,037 individuals
             born in Dunedin, New Zealand, in 1972-1973 and followed to
             age 45, with 94% retention. Cannabis use and dependence were
             assessed at ages 18, 21, 26, 32, 38, and 45. IQ was assessed
             at ages 7, 9, 11, and 45. Specific neuropsychological
             functions and hippocampal volume were assessed at age
             45.<h4>Results</h4>Long-term cannabis users showed IQ
             decline from childhood to midlife (mean=-5.5 IQ points),
             poorer learning and processing speed relative to their
             childhood IQ, and informant-reported memory and attention
             problems. These deficits were specific to long-term cannabis
             users because they were either not present or were smaller
             among long-term tobacco users, long-term alcohol users,
             midlife recreational cannabis users, and cannabis quitters.
             Cognitive deficits among long-term cannabis users could not
             be explained by persistent tobacco, alcohol, or other
             illicit drug use, childhood socioeconomic status, low
             childhood self-control, or family history of substance
             dependence. Long-term cannabis users showed smaller
             hippocampal volume, but smaller hippocampal volume did not
             statistically mediate cannabis-related cognitive
             deficits.<h4>Conclusions</h4>Long-term cannabis users showed
             cognitive deficits and smaller hippocampal volume in
             midlife. Research is needed to ascertain whether long-term
             cannabis users show elevated rates of dementia in later
             life.},
   Doi = {10.1176/appi.ajp.2021.21060664},
   Key = {fds364143}
}

@article{fds364171,
   Author = {Bourassa, KJ and Moffitt, TE and Ambler, A and Hariri, AR and Harrington, H and Houts, RM and Ireland, D and Knodt, A and Poulton, R and Ramrakha, S and Caspi, A},
   Title = {Association of Treatable Health Conditions During
             Adolescence With Accelerated Aging at Midlife.},
   Journal = {JAMA pediatrics},
   Volume = {176},
   Number = {4},
   Pages = {392-399},
   Year = {2022},
   Month = {April},
   Abstract = {<h4>Importance</h4>Biological aging is a distinct construct
             from health; however, people who age quickly are more likely
             to experience poor health. Identifying pediatric health
             conditions associated with accelerated aging could help
             develop treatment approaches to slow midlife aging and
             prevent poor health in later life.<h4>Objective</h4>To
             examine the association between 4 treatable health
             conditions in adolescence and accelerated aging at
             midlife.<h4>Design, setting, and participants</h4>This
             cohort study analyzed data from participants in the Dunedin
             Study, a longitudinal investigation of health and behavior
             among a birth cohort born between April 1, 1972, and March
             31, 1973, in Dunedin, New Zealand, and followed up until age
             45 years. Participants underwent an assessment at age 45
             years and had data for at least 1 adolescent health
             condition (asthma, smoking, obesity, and psychological
             disorders) and outcome measure (pace of aging, gait speed,
             brain age, and facial age). Data analysis was performed from
             February 11 to September 27, 2021.<h4>Exposures</h4>Asthma,
             cigarette smoking, obesity, and psychological disorders were
             assessed at age 11, 13, and 15 years.<h4>Main outcomes and
             measures</h4>The outcome was a midlife aging factor
             composite score comprising 4 measures of biological aging:
             pace of aging, gait speed, brain age (specifically, BrainAGE
             score), and facial age.<h4>Results</h4>A total of 910
             participants (459 men [50.4%]) met the inclusion criteria,
             including an assessment at age 45 years. Participants who
             had smoked daily (0.61 [95% CI, 0.43-0.79] SD units), had
             obesity (0.82 [95% CI, 0.59-1.06] SD units), or had a
             psychological disorder diagnosis (0.43 [95% CI, 0.29-0.56]
             SD units) during adolescence were biologically older at
             midlife compared with participants without these conditions.
             Participants with asthma were not biologically older at
             midlife (0.02 [95% CI, -0.14 to 0.19] SD units) compared
             with those without asthma. These results remained unchanged
             after adjusting for childhood risk factors such as poor
             health, socioeconomic disadvantage, and adverse
             experiences.<h4>Conclusions and relevance</h4>This study
             found that adolescent smoking, obesity, and psychological
             disorder diagnoses were associated with older biological age
             at midlife. These health conditions could be treated during
             adolescence to reduce the risk of accelerated biological
             aging later in life.},
   Doi = {10.1001/jamapediatrics.2021.6417},
   Key = {fds364171}
}

@article{fds364966,
   Author = {Kragel, PA and Hariri, AR and LaBar, KS},
   Title = {The Temporal Dynamics of Spontaneous Emotional Brain States
             and Their Implications for Mental Health.},
   Journal = {Journal of cognitive neuroscience},
   Volume = {34},
   Number = {5},
   Pages = {715-728},
   Year = {2022},
   Month = {March},
   Abstract = {Temporal processes play an important role in elaborating and
             regulating emotional responding during routine mind
             wandering. However, it is unknown whether the human brain
             reliably transitions among multiple emotional states at rest
             and how psychopathology alters these affect dynamics. Here,
             we combined pattern classification and stochastic process
             modeling to investigate the chronometry of spontaneous brain
             activity indicative of six emotions (anger, contentment,
             fear, happiness, sadness, and surprise) and a neutral state.
             We modeled the dynamic emergence of these brain states
             during resting-state fMRI and validated the results across
             two population cohorts-the Duke Neurogenetics Study and the
             Nathan Kline Institute Rockland Sample. Our findings
             indicate that intrinsic emotional brain dynamics are
             effectively characterized as a discrete-time Markov process,
             with affective states organized around a neutral hub. The
             centrality of this network hub is disrupted in individuals
             with psychopathology, whose brain state transitions exhibit
             greater inertia and less frequent resetting from emotional
             to neutral states. These results yield novel insights into
             how the brain signals spontaneous emotions and how
             alterations in their temporal dynamics contribute to
             compromised mental health.},
   Doi = {10.1162/jocn_a_01787},
   Key = {fds364966}
}

@article{fds368528,
   Author = {Reuben, A and Moffitt, TE and Abraham, WC and Ambler, A and Elliott, ML and Hariri, AR and Harrington, H and Hogan, S and Houts, RM and Ireland, D and Knodt, AR and Leung, J and Pearson, A and Poulton, R and Purdy, SC and Ramrakha, S and Rasmussen, LJH and Sugden, K and Thorne, PR and Williams, B and Wilson, G and Caspi, A},
   Title = {Improving risk indexes for Alzheimer's disease and related
             dementias for use in midlife.},
   Journal = {Brain communications},
   Volume = {4},
   Number = {5},
   Pages = {fcac223},
   Year = {2022},
   Month = {January},
   Abstract = {Knowledge of a person's risk for Alzheimer's disease and
             related dementias (ADRDs) is required to triage candidates
             for preventive interventions, surveillance, and treatment
             trials. ADRD risk indexes exist for this purpose, but each
             includes only a subset of known risk factors. Information
             missing from published indexes could improve risk
             prediction. In the Dunedin Study of a population-representative
             New Zealand-based birth cohort followed to midlife
             (<i>N</i> = 938, 49.5% female), we compared associations
             of four leading risk indexes with midlife antecedents of
             ADRD against a novel benchmark index comprised of nearly all
             known ADRD risk factors, the Dunedin ADRD Risk Benchmark
             (DunedinARB). Existing indexes included the Cardiovascular
             Risk Factors, Aging, and Dementia index (CAIDE), LIfestyle
             for BRAin health index (LIBRA), Australian National
             University Alzheimer's Disease Risk Index (ANU-ADRI), and
             risks selected by the Lancet Commission on Dementia. The
             Dunedin benchmark was comprised of 48 separate indicators of
             risk organized into 10 conceptually distinct risk domains.
             Midlife antecedents of ADRD treated as outcome measures
             included age-45 measures of brain structural integrity
             [magnetic resonance imaging-assessed: (i)
             machine-learning-algorithm-estimated brain age, (ii)
             log-transformed volume of white matter hyperintensities, and
             (iii) mean grey matter volume of the hippocampus] and
             measures of brain functional integrity [(i) objective
             cognitive function assessed via the Wechsler Adult
             Intelligence Scale-IV, (ii) subjective problems in everyday
             cognitive function, and (iii) objective cognitive decline
             measured as residualized change in cognitive scores from
             childhood to midlife on matched Weschler Intelligence
             scales]. All indexes were quantitatively distributed and
             proved informative about midlife antecedents of ADRD,
             including algorithm-estimated brain age (<i>β</i>'s from
             0.16 to 0.22), white matter hyperintensities volume
             (<i>β</i>'s from 0.16 to 0.19), hippocampal volume
             (<i>β</i>'s from -0.08 to -0.11), tested cognitive deficits
             (<i>β</i>'s from -0.36 to -0.49), everyday cognitive
             problems (<i>β</i>'s from 0.14 to 0.38), and longitudinal
             cognitive decline (<i>β</i>'s from -0.18 to -0.26).
             Existing indexes compared favourably to the comprehensive
             benchmark in their association with the brain structural
             integrity measures but were outperformed in their
             association with the functional integrity measures,
             particularly subjective cognitive problems and tested
             cognitive decline. Results indicated that existing indexes
             could be improved with targeted additions, particularly of
             measures assessing socioeconomic status, physical and
             sensory function, epigenetic aging, and subjective overall
             health. Existing premorbid ADRD risk indexes perform well in
             identifying linear gradients of risk among members of the
             general population at midlife, even when they include only a
             small subset of potential risk factors. They could be
             improved, however, with targeted additions to more
             holistically capture the different facets of risk for this
             multiply determined, age-related disease.},
   Doi = {10.1093/braincomms/fcac223},
   Key = {fds368528}
}

@article{fds364215,
   Author = {Miles, AE and Dos Santos and FC and Byrne, EM and Renteria, ME and McIntosh, AM and Adams, MJ and Pistis, G and Castelao, E and Preisig, M and Baune, BT and Schubert, KO and Lewis, CM and Jones, LA and Jones, I and Uher, R and Smoller, JW and Perlis, RH and Levinson, DF and Potash, JB and Weissman, MM and Shi, J and Lewis, G and Penninx, BWJH and Boomsma, DI and Hamilton, SP and Major Depressive Disorder Working Group of the
             Psychiatric Genomics Consortium, and Sibille, E and Hariri, AR and Nikolova, YS},
   Title = {Transcriptome-based polygenic score links depression-related
             corticolimbic gene expression changes to sex-specific brain
             morphology and depression risk.},
   Journal = {Neuropsychopharmacology : official publication of the
             American College of Neuropsychopharmacology},
   Volume = {46},
   Number = {13},
   Pages = {2304-2311},
   Year = {2021},
   Month = {December},
   Abstract = {Studies in post-mortem human brain tissue have associated
             major depressive disorder (MDD) with cortical transcriptomic
             changes, whose potential in vivo impact remains unexplored.
             To address this translational gap, we recently developed a
             transcriptome-based polygenic risk score (T-PRS) based on
             common functional variants capturing 'depression-like'
             shifts in cortical gene expression. Here, we used a
             non-clinical sample of young adults (n = 482, Duke
             Neurogenetics Study: 53% women; aged 19.8 ± 1.2 years)
             to map T-PRS onto brain morphology measures, including
             Freesurfer-derived subcortical volume, cortical thickness,
             surface area, and local gyrification index, as well as broad
             MDD risk, indexed by self-reported family history of
             depression. We conducted side-by-side comparisons with a PRS
             independently derived from a Psychiatric Genomics Consortium
             (PGC) MDD GWAS (PGC-PRS), and sought to link T-PRS with
             diagnosis and symptom severity directly in PGC-MDD
             participants (n = 29,340, 59% women; 12,923 MDD cases,
             16,417 controls). T-PRS was associated with smaller amygdala
             volume in women (t = -3.478, p = 0.001) and lower
             prefrontal gyrification across sexes. In men, T-PRS was
             associated with hypergyrification in temporal and occipital
             regions. Prefrontal hypogyrification mediated a
             male-specific indirect link between T-PRS and familial
             depression (b = 0.005, p = 0.029). PGC-PRS was
             similarly associated with lower amygdala volume and cortical
             gyrification; however, both effects were male-specific and
             hypogyrification emerged in distinct parietal and
             temporo-occipital regions, unassociated with familial
             depression. In PGC-MDD, T-PRS did not predict diagnosis
             (OR = 1.007, 95% CI = [0.997-1.018]) but correlated
             with symptom severity in men (rho = 0.175,
             p = 7.957 × 10<sup>-4</sup>) in one cohort
             (N = 762, 48% men). Depression-like shifts in cortical
             gene expression have sex-specific effects on brain
             morphology and may contribute to broad depression
             vulnerability in men.},
   Doi = {10.1038/s41386-021-01189-x},
   Key = {fds364215}
}

@article{fds368901,
   Author = {Carlisi, CO and Moffitt, TE and Knodt, AR and Harrington, H and Langevin, S and Ireland, D and Melzer, TR and Poulton, R and Ramrakha,
             S and Caspi, A and Hariri, AR and Viding, E},
   Title = {Association of subcortical gray-matter volumes with
             life-course-persistent antisocial behavior in a
             population-representative longitudinal birth
             cohort.},
   Journal = {Development and psychopathology},
   Pages = {1-11},
   Year = {2021},
   Month = {October},
   Abstract = {Neuropsychological evidence supports the developmental
             taxonomy theory of antisocial behavior, suggesting that
             abnormal brain development distinguishes
             life-course-persistent from adolescence-limited antisocial
             behavior. Recent neuroimaging work confirmed that
             prospectively-measured life-course-persistent antisocial
             behavior is associated with differences in cortical brain
             structure. Whether this extends to subcortical brain
             structures remains uninvestigated. This study compared
             subcortical gray-matter volumes between 672 members of the
             Dunedin Study previously defined as exhibiting
             life-course-persistent, adolescence-limited or low-level
             antisocial behavior based on repeated assessments at ages
             7-26 years. Gray-matter volumes of 10 subcortical structures
             were compared across groups. The life-course-persistent
             group had lower volumes of amygdala, brain stem, cerebellum,
             hippocampus, pallidum, thalamus, and ventral diencephalon
             compared to the low-antisocial group. Differences between
             life-course-persistent and adolescence-limited individuals
             were comparable in effect size to differences between
             life-course-persistent and low-antisocial individuals, but
             were not statistically significant due to less statistical
             power. Gray-matter volumes in adolescence-limited
             individuals were near the norm in this population-representative
             cohort and similar to volumes in low-antisocial individuals.
             Although this study could not establish causal links between
             brain volume and antisocial behavior, it constitutes new
             biological evidence that all people with antisocial behavior
             are not the same, supporting a need for greater
             developmental and diagnostic precision in clinical,
             forensic, and policy-based interventions.},
   Doi = {10.1017/s0954579421000377},
   Key = {fds368901}
}

@article{fds364144,
   Author = {Lam, M and Chen, C-Y and Ge, T and Xia, Y and Hill, DW and Trampush, JW and Yu, J and Knowles, E and Davies, G and Stahl, EA and Huckins, L and Liewald, DC and Djurovic, S and Melle, I and Christoforou, A and Reinvang, I and DeRosse, P and Lundervold, AJ and Steen, VM and Espeseth, T and Räikkönen, K and Widen, E and Palotie, A and Eriksson,
             JG and Giegling, I and Konte, B and Hartmann, AM and Roussos, P and Giakoumaki, S and Burdick, KE and Payton, A and Ollier, W and Chiba-Falek, O and Koltai, DC and Need, AC and Cirulli, ET and Voineskos, AN and Stefanis, NC and Avramopoulos, D and Hatzimanolis,
             A and Smyrnis, N and Bilder, RM and Freimer, NB and Cannon, TD and London,
             E and Poldrack, RA and Sabb, FW and Congdon, E and Conley, ED and Scult,
             MA and Dickinson, D and Straub, RE and Donohoe, G and Morris, D and Corvin,
             A and Gill, M and Hariri, AR and Weinberger, DR and Pendleton, N and Bitsios, P and Rujescu, D and Lahti, J and Le Hellard and S and Keller, MC and Andreassen, OA and Deary, IJ and Glahn, DC and Huang, H and Liu, C and Malhotra, AK and Lencz, T},
   Title = {Identifying nootropic drug targets via large-scale cognitive
             GWAS and transcriptomics.},
   Journal = {Neuropsychopharmacology},
   Volume = {46},
   Number = {10},
   Pages = {1788-1801},
   Year = {2021},
   Month = {September},
   Abstract = {Broad-based cognitive deficits are an enduring and disabling
             symptom for many patients with severe mental illness, and
             these impairments are inadequately addressed by current
             medications. While novel drug targets for schizophrenia and
             depression have emerged from recent large-scale genome-wide
             association studies (GWAS) of these psychiatric disorders,
             GWAS of general cognitive ability can suggest potential
             targets for nootropic drug repurposing. Here, we (1)
             meta-analyze results from two recent cognitive GWAS to
             further enhance power for locus discovery; (2) employ
             several complementary transcriptomic methods to identify
             genes in these loci that are credibly associated with
             cognition; and (3) further annotate the resulting genes
             using multiple chemoinformatic databases to identify
             "druggable" targets. Using our meta-analytic data set
             (N = 373,617), we identified 241 independent
             cognition-associated loci (29 novel), and 76 genes were
             identified by 2 or more methods of gene identification.
             Actin and chromatin binding gene sets were identified as
             novel pathways that could be targeted via drug repurposing.
             Leveraging our transcriptomic and chemoinformatic databases,
             we identified 16 putative genes targeted by existing drugs
             potentially available for cognitive repurposing.},
   Doi = {10.1038/s41386-021-01023-4},
   Key = {fds364144}
}

@article{fds364145,
   Author = {Elliott, ML and Knodt, AR and Hariri, AR},
   Title = {Striving toward translation: strategies for reliable fMRI
             measurement.},
   Journal = {Trends in cognitive sciences},
   Volume = {25},
   Number = {9},
   Pages = {776-787},
   Year = {2021},
   Month = {September},
   Abstract = {fMRI has considerable potential as a translational tool for
             understanding risk, prioritizing interventions, and
             improving the treatment of brain disorders. However, recent
             studies have found that many of the most widely used fMRI
             measures have low reliability, undermining this potential.
             Here, we argue that many fMRI measures are unreliable
             because they were designed to identify group effects, not to
             precisely quantify individual differences. We then highlight
             four emerging strategies [extended aggregation, reliability
             modeling, multi-echo fMRI (ME-fMRI), and stimulus design]
             that build on established psychometric properties to
             generate more precise and reliable fMRI measures. By
             adopting such strategies to improve reliability, we are
             optimistic that fMRI can fulfill its potential as a clinical
             tool.},
   Doi = {10.1016/j.tics.2021.05.008},
   Key = {fds364145}
}

@article{fds347350,
   Author = {Romer, AL and Knodt, AR and Sison, ML and Ireland, D and Houts, R and Ramrakha, S and Poulton, R and Keenan, R and Melzer, TR and Moffitt, TE and Caspi, A and Hariri, AR},
   Title = {Replicability of structural brain alterations associated
             with general psychopathology: evidence from a
             population-representative birth cohort.},
   Journal = {Molecular psychiatry},
   Volume = {26},
   Number = {8},
   Pages = {3839-3846},
   Year = {2021},
   Month = {August},
   Abstract = {Transdiagnostic research has identified a general
             psychopathology factor-often called the 'p' factor-that
             accounts for shared variation across internalizing,
             externalizing, and thought disorders in diverse samples. It
             has been argued that the p factor may reflect dysfunctional
             thinking present in serious mental illness. In support of
             this, we previously used a theory-free, data-driven
             multimodal neuroimaging approach to find that higher p
             factor scores are associated with structural alterations
             within a cerebello-thalamo-cortical circuit (CTCC) and
             visual association cortex, both of which are important for
             monitoring and coordinating information processing in the
             service of executive control. Here we attempt to replicate
             these associations by conducting region-of-interest analyses
             using data from 875 members of the Dunedin Longitudinal
             Study, a five-decade study of a population-representative
             birth cohort, collected when they were 45 years old. We
             further sought to replicate a more recent report that p
             factor scores can be predicted by patterns of distributed
             cerebellar morphology as estimated through independent
             component analysis. We successfully replicated associations
             between higher p factor scores and both reduced gray matter
             volume of the visual association cortex and fractional
             anisotropy of pontine white matter pathways within the CTCC.
             In contrast, we failed to replicate prior associations
             between cerebellar structure and p factor scores.
             Collectively, our findings encourage further focus on the
             CTCC and visual association cortex as core neural substrates
             and potential biomarkers of general psychopathology.},
   Doi = {10.1038/s41380-019-0621-z},
   Key = {fds347350}
}

@article{fds347349,
   Author = {Elliott, ML and Belsky, DW and Knodt, AR and Ireland, D and Melzer, TR and Poulton, R and Ramrakha, S and Caspi, A and Moffitt, TE and Hariri,
             AR},
   Title = {Brain-age in midlife is associated with accelerated
             biological aging and cognitive decline in a longitudinal
             birth cohort.},
   Journal = {Molecular psychiatry},
   Volume = {26},
   Number = {8},
   Pages = {3829-3838},
   Year = {2021},
   Month = {August},
   Abstract = {An individual's brainAGE is the difference between
             chronological age and age predicted from machine-learning
             models of brain-imaging data. BrainAGE has been proposed as
             a biomarker of age-related deterioration of the brain.
             Having an older brainAGE has been linked to Alzheimer's,
             dementia, and mortality. However, these findings are largely
             based on cross-sectional associations which can confuse age
             differences with cohort differences. To illuminate the
             validity of brainAGE as a biomarker of accelerated brain
             aging, a study is needed of a large cohort all born in the
             same year who nevertheless vary on brainAGE. In the Dunedin
             Study, a population-representative 1972-73 birth cohort, we
             measured brainAGE at age 45 years, as well as the pace of
             biological aging and cognitive decline in longitudinal data
             from childhood to midlife (N = 869). In this cohort, all
             chronological age 45 years, brainAGE was measured reliably
             (ICC = 0.81) and ranged from 24 to 72 years. Those with
             older midlife brainAGEs tended to have poorer cognitive
             function in both adulthood and childhood, as well as
             impaired brain health at age 3. Furthermore, those with
             older brainAGEs had an accelerated pace of biological aging,
             older facial appearance, and early signs of cognitive
             decline from childhood to midlife. These findings help to
             validate brainAGE as a potential surrogate biomarker for
             midlife intervention studies that seek to measure
             dementia-prevention efforts in midlife. However, the
             findings also caution against the assumption that brainAGE
             scores represent only age-related deterioration of the brain
             as they may also index central nervous system variation
             present since childhood.},
   Doi = {10.1038/s41380-019-0626-7},
   Key = {fds347349}
}

@article{fds356131,
   Author = {Gehred, MZ and Knodt, AR and Ambler, A and Bourassa, KJ and Danese, A and Elliott, ML and Hogan, S and Ireland, D and Poulton, R and Ramrakha, S and Reuben, A and Sison, ML and Moffitt, TE and Hariri, AR and Caspi,
             A},
   Title = {Long-term Neural Embedding of Childhood Adversity in a
             Population-Representative Birth Cohort Followed for 5
             Decades.},
   Journal = {Biological psychiatry},
   Volume = {90},
   Number = {3},
   Pages = {182-193},
   Year = {2021},
   Month = {August},
   Abstract = {<h4>Background</h4>Childhood adversity has been previously
             associated with alterations in brain structure, but
             heterogeneous designs, methods, and measures have
             contributed to mixed results and have impeded progress in
             mapping the biological embedding of childhood adversity. We
             sought to identify long-term differences in structural brain
             integrity associated with childhood adversity.<h4>Methods</h4>Multiple
             regression was used to test associations between
             prospectively ascertained adversity during childhood and
             adversity retrospectively reported in adulthood with
             structural magnetic resonance imaging measures of midlife
             global and regional cortical thickness, cortical surface
             area, and subcortical gray matter volume in 861 (425 female)
             members of the Dunedin Study, a longitudinal investigation
             of a population-representative birth cohort.<h4>Results</h4>Both
             prospectively ascertained childhood adversity and
             retrospectively reported adversity were associated with
             alterations in midlife structural brain integrity, but
             associations with prospectively ascertained childhood
             adversity were consistently stronger and more widely
             distributed than associations with retrospectively reported
             childhood adversity. Sensitivity analyses revealed that
             these associations were not driven by any particular
             adversity or category of adversity (i.e., threat or
             deprivation) or by childhood socioeconomic disadvantage.
             Network enrichment analyses revealed that these associations
             were not localized but were broadly distributed along a
             hierarchical cortical gradient of information
             processing.<h4>Conclusions</h4>Exposure to childhood
             adversity broadly is associated with widespread differences
             in midlife gray matter across cortical and subcortical
             structures, suggesting that biological embedding of
             childhood adversity in the brain is long lasting, but not
             localized. Research using retrospectively reported adversity
             likely underestimates the magnitude of these associations.
             These findings may inform future research investigating
             mechanisms through which adversity becomes embedded in the
             brain and influences mental health and cognition.},
   Doi = {10.1016/j.biopsych.2021.02.971},
   Key = {fds356131}
}

@article{fds364967,
   Author = {Romer, AL and Hariri, AR and Strauman, TJ},
   Title = {Regulatory focus and the p factor: Evidence for
             self-regulatory dysfunction as a transdiagnostic feature of
             general psychopathology.},
   Journal = {Journal of psychiatric research},
   Volume = {137},
   Pages = {178-185},
   Year = {2021},
   Month = {May},
   Abstract = {A general psychopathology ('p') factor captures
             transdiagnostic features of mental illness; however, the
             meaning of the p factor remains unclear. Regulatory focus
             theory postulates that individuals regulate goal pursuit
             either by maximizing gains (promotion) or minimizing losses
             (prevention). As maladaptive goal pursuit has been
             associated with multiple categorical disorders, we examined
             whether individual differences in promotion and prevention
             goal pursuit are associated with p as well as internalizing-
             and externalizing-specific factors using structural equation
             modeling of data from 1330 volunteers aged 18-22.
             Unsuccessful attainment of promotion and prevention goals
             was related to increased levels of p. Over and above
             relations with the p factor, unsuccessful attainment of
             promotion goals was associated with higher
             internalizing-specific psychopathology, whereas unsuccessful
             attainment of prevention goals was related to higher
             externalizing-specific psychopathology. These associations
             also were separable from related personality traits. After
             controlling for sex differences in the composition of the
             psychopathology factors, there were no sex differences in
             the relations between promotion and prevention goal pursuit
             and p and specific internalizing and externalizing factors.
             These findings suggest higher general psychopathology
             reflects poorer overall self-regulation of goal pursuit and
             that maladaptive promotion and prevention orientations also
             are associated with internalizing- and externalizing-specific
             psychopathology, respectively.},
   Doi = {10.1016/j.jpsychires.2021.02.051},
   Key = {fds364967}
}

@article{fds364968,
   Author = {Elliott, ML and Knodt, AR and Caspi, A and Moffitt, TE and Hariri,
             AR},
   Title = {Need for Psychometric Theory in Neuroscience Research and
             Training: Reply to Kragel et al. (2021).},
   Journal = {Psychological science},
   Volume = {32},
   Number = {4},
   Pages = {627-629},
   Year = {2021},
   Month = {April},
   Doi = {10.1177/0956797621996665},
   Key = {fds364968}
}

@article{fds364216,
   Author = {Pfeiffer, JR and Bustamante, AC and Kim, GS and Armstrong, D and Knodt,
             AR and Koenen, KC and Hariri, AR and Uddin, M},
   Title = {Associations between childhood family emotional health,
             fronto-limbic grey matter volume, and saliva 5mC in young
             adulthood.},
   Journal = {Clinical epigenetics},
   Volume = {13},
   Number = {1},
   Pages = {68},
   Year = {2021},
   Month = {March},
   Abstract = {<h4>Background</h4>Poor family emotional health (FEH) during
             childhood is prevalent and impactful, and likely confers
             similar neurodevelopmental risks as other adverse social
             environments. Pointed FEH study efforts are underdeveloped,
             and the mechanisms by which poor FEH are biologically
             embedded are unclear. The current exploratory study examined
             whether variability in 5-methyl-cytosine (5mC) and
             fronto-limbic grey matter volume may represent pathways
             through which FEH may become biologically
             embedded.<h4>Results</h4>In 98 university students aged
             18-22 years, retrospective self-reported childhood FEH was
             associated with right hemisphere hippocampus (b = 10.4,
             p = 0.005), left hemisphere amygdala (b = 5.3,
             p = 0.009), and right hemisphere amygdala (b = 5.8,
             p = 0.016) volumes. After pre-processing and filtering
             to 5mC probes correlated between saliva and brain, analyses
             showed that childhood FEH was associated with 49 5mC
             principal components (module eigengenes; MEs)
             (p<sub>range</sub> = 3 × 10<sup>-6</sup> to 0.047).
             Saliva-derived 5mC MEs partially mediated the association
             between FEH and right hippocampal volume (Burlywood ME
             indirect effect b = - 111, p = 0.014), and fully
             mediated the FEH and right amygdala volume relationship
             (Pink4 ME indirect effect b = - 48, p = 0.026).
             Modules were enriched with probes falling in genes with
             immune, central nervous system (CNS), cellular
             development/differentiation, and metabolic
             functions.<h4>Conclusions</h4>Findings extend work
             highlighting neurodevelopmental variability associated with
             adverse social environment exposure during childhood by
             specifically implicating poor FEH, while informing a
             mechanism of biological embedding. FEH-associated epigenetic
             signatures could function as proxies of altered
             fronto-limbic grey matter volume associated with poor
             childhood FEH and inform further investigation into
             primarily affected tissues such as endocrine, immune, and
             CNS cell types.},
   Doi = {10.1186/s13148-021-01056-y},
   Key = {fds364216}
}

@article{fds364162,
   Author = {Elliott, ML and Caspi, A and Houts, RM and Ambler, A and Broadbent, JM and Hancox, RJ and Harrington, H and Hogan, S and Keenan, R and Knodt, A and Leung, JH and Melzer, TR and Purdy, SC and Ramrakha, S and Richmond-Rakerd, LS and Righarts, A and Sugden, K and Thomson, WM and Thorne, PR and Williams, BS and Wilson, G and Hariri, AR and Poulton, R and Moffitt, TE},
   Title = {Disparities in the pace of biological aging among midlife
             adults of the same chronological age have implications for
             future frailty risk and policy.},
   Journal = {Nature aging},
   Volume = {1},
   Number = {3},
   Pages = {295-308},
   Year = {2021},
   Month = {March},
   Abstract = {Some humans age faster than others. Variation in biological
             aging can be measured in midlife, but the implications of
             this variation are poorly understood. We tested associations
             between midlife biological aging and indicators of future
             frailty-risk in the Dunedin cohort of 1037 infants born the
             same year and followed to age 45. Participants' Pace of
             Aging was quantified by tracking declining function in 19
             biomarkers indexing the cardiovascular, metabolic, renal,
             immune, dental, and pulmonary systems across ages 26, 32,
             38, and 45 years. At age 45 in 2019, participants with
             faster Pace of Aging had more cognitive difficulties, signs
             of advanced brain aging, diminished sensory-motor functions,
             older appearance, and more pessimistic perceptions of aging.
             People who are aging more rapidly than same-age peers in
             midlife may prematurely need supports to sustain
             independence that are usually reserved for older adults.
             Chronological age does not adequately identify need for such
             supports.},
   Doi = {10.1038/s43587-021-00044-4},
   Key = {fds364162}
}

@article{fds364217,
   Author = {Mareckova, K and Hawco, C and Santos, FCD and Bakht, A and Calarco, N and Miles, AE and Voineskos, AN and Sibille, E and Hariri, AR and Nikolova,
             YS},
   Title = {Correction: Novel polygenic risk score as a translational
             tool linking depression-related changes in the corticolimbic
             transcriptome with neural face processing and anhedonic
             symptoms.},
   Journal = {Translational psychiatry},
   Volume = {11},
   Number = {1},
   Pages = {152},
   Year = {2021},
   Month = {March},
   Doi = {10.1038/s41398-021-01277-y},
   Key = {fds364217}
}

@article{fds364969,
   Author = {Romer, AL and Elliott, ML and Knodt, AR and Sison, ML and Ireland, D and Houts, R and Ramrakha, S and Poulton, R and Keenan, R and Melzer, TR and Moffitt, TE and Caspi, A and Hariri, AR},
   Title = {Pervasively Thinner Neocortex as a Transdiagnostic Feature
             of General Psychopathology.},
   Journal = {The American journal of psychiatry},
   Volume = {178},
   Number = {2},
   Pages = {174-182},
   Year = {2021},
   Month = {February},
   Abstract = {<h4>Objective</h4>Neuroimaging research has revealed that
             structural brain alterations are common across broad
             diagnostic families of disorders rather than specific to a
             single psychiatric disorder. Such overlap in the structural
             brain correlates of mental disorders mirrors already
             well-documented phenotypic comorbidity of psychiatric
             symptoms and diagnoses, which can be indexed by a general
             psychopathology or <i>p</i> factor. The authors hypothesized
             that if general psychopathology drives the convergence of
             structural alterations common across disorders, then 1)
             there should be few associations unique to any one
             diagnostic family of disorders, and 2) associations with the
             <i>p</i> factor should overlap with those for the broader
             diagnostic families.<h4>Methods</h4>Analyses were conducted
             on structural MRI and psychopathology data collected from
             861 members of the population-representative Dunedin
             Multidisciplinary Health and Development Study at age
             45.<h4>Results</h4>Study members with high scores across
             three broad diagnostic families of disorders (externalizing,
             internalizing, thought disorder) exhibited highly
             overlapping patterns of reduced global and widely
             distributed parcel-wise neocortical thickness. Study members
             with high <i>p</i> factor scores exhibited patterns of
             reduced global and parcel-wise neocortical thickness nearly
             identical to those associated with the three broad
             diagnostic families.<h4>Conclusions</h4>A pattern of
             pervasively reduced neocortical thickness appears to be
             common across all forms of mental disorders and may
             represent a transdiagnostic feature of general
             psychopathology. As has been documented with regard to
             symptoms and diagnoses, the underlying brain structural
             correlates of mental disorders may not exhibit specificity,
             and the continued pursuit of such specific correlates may
             limit progress toward more effective strategies for
             etiological understanding, prevention, and
             intervention.},
   Doi = {10.1176/appi.ajp.2020.19090934},
   Key = {fds364969}
}

@article{fds364181,
   Author = {Richmond-Rakerd, LS and Caspi, A and Ambler, A and d'Arbeloff, T and de
             Bruine, M and Elliott, M and Harrington, H and Hogan, S and Houts, RM and Ireland, D and Keenan, R and Knodt, AR and Melzer, TR and Park, S and Poulton, R and Ramrakha, S and Rasmussen, LJH and Sack, E and Schmidt,
             AT and Sison, ML and Wertz, J and Hariri, AR and Moffitt,
             TE},
   Title = {Childhood self-control forecasts the pace of midlife aging
             and preparedness for old age.},
   Journal = {Proceedings of the National Academy of Sciences of the
             United States of America},
   Volume = {118},
   Number = {3},
   Pages = {e2010211118},
   Year = {2021},
   Month = {January},
   Abstract = {The ability to control one's own emotions, thoughts, and
             behaviors in early life predicts a range of positive
             outcomes in later life, including longevity. Does it also
             predict how well people age? We studied the association
             between self-control and midlife aging in a
             population-representative cohort of children followed from
             birth to age 45 y, the Dunedin Study. We measured children's
             self-control across their first decade of life using a
             multi-occasion/multi-informant strategy. We measured their
             pace of aging and aging preparedness in midlife using
             measures derived from biological and physiological
             assessments, structural brain-imaging scans, observer
             ratings, self-reports, informant reports, and administrative
             records. As adults, children with better self-control aged
             more slowly in their bodies and showed fewer signs of aging
             in their brains. By midlife, these children were also better
             equipped to manage a range of later-life health, financial,
             and social demands. Associations with children's
             self-control could be separated from their social class
             origins and intelligence, indicating that self-control might
             be an active ingredient in healthy aging. Children also
             shifted naturally in their level of self-control across
             adult life, suggesting the possibility that self-control may
             be a malleable target for intervention. Furthermore,
             individuals' self-control in adulthood was associated with
             their aging outcomes after accounting for their self-control
             in childhood, indicating that midlife might offer another
             window of opportunity to promote healthy
             aging.},
   Doi = {10.1073/pnas.2010211118},
   Key = {fds364181}
}

@article{fds364146,
   Author = {d'Arbeloff, T and Elliott, ML and Knodt, AR and Sison, M and Melzer, TR and Ireland, D and Ramrakha, S and Poulton, R and Caspi, A and Moffitt, TE and Hariri, AR},
   Title = {Midlife Cardiovascular Fitness Is Reflected in the Brain's
             White Matter.},
   Journal = {Frontiers in aging neuroscience},
   Volume = {13},
   Pages = {652575},
   Year = {2021},
   Month = {January},
   Abstract = {Disappointing results from clinical trials designed to delay
             structural brain decline and the accompanying increase in
             risk for dementia in older adults have precipitated a shift
             in testing promising interventions from late in life toward
             midlife before irreversible damage has accumulated. This
             shift, however, requires targeting midlife biomarkers that
             are associated with clinical changes manifesting only in
             late life. Here we explored possible links between one
             putative biomarker, distributed integrity of brain white
             matter, and two intervention targets, cardiovascular fitness
             and healthy lifestyle behaviors, in midlife. At age 45,
             fractional anisotropy (FA) derived from diffusion weighted
             MRI was used to estimate the microstructural integrity of
             distributed white matter tracts in a population-representative
             birth cohort. Age-45 cardiovascular fitness
             (VO<sub>2</sub>Max; <i>N</i> = 801) was estimated from heart
             rates obtained during submaximal exercise tests; age-45
             healthy lifestyle behaviors were estimated using the Nyberg
             Health Index (<i>N</i> = 854). Ten-fold cross-validated
             elastic net predictive modeling revealed that estimated
             VO<sub>2</sub>Max was modestly associated with distributed
             FA. In contrast, there was no significant association
             between Nyberg Health Index scores and FA. Our findings
             suggest that cardiovascular fitness levels, but not healthy
             lifestyle behaviors, are associated with the distributed
             integrity of white matter in the brain in midlife. These
             patterns could help inform future clinical intervention
             research targeting ADRDs.},
   Doi = {10.3389/fnagi.2021.652575},
   Key = {fds364146}
}

@article{fds364970,
   Author = {Rasmussen, LJH and Caspi, A and Ambler, A and Danese, A and Elliott, M and Eugen-Olsen, J and Hariri, AR and Harrington, H and Houts, R and Poulton, R and Ramrakha, S and Sugden, K and Williams, B and Moffitt,
             TE},
   Title = {Association Between Elevated suPAR, a New Biomarker of
             Inflammation, and Accelerated Aging.},
   Journal = {The journals of gerontology. Series A, Biological sciences
             and medical sciences},
   Volume = {76},
   Number = {2},
   Pages = {318-327},
   Year = {2021},
   Month = {January},
   Abstract = {<h4>Background</h4>To understand and measure the association
             between chronic inflammation, aging, and age-related
             diseases, broadly applicable standard biomarkers of systemic
             chronic inflammation are needed. We tested whether elevated
             blood levels of the emerging chronic inflammation marker
             soluble urokinase plasminogen activator receptor (suPAR)
             were associated with accelerated aging, lower functional
             capacity, and cognitive decline.<h4>Methods</h4>We used data
             from the Dunedin Study, a population-representative
             1972-1973 New Zealand birth cohort (n = 1037) that has
             observed participants to age 45 years. Plasma suPAR levels
             were analyzed at ages 38 and 45 years. We performed
             regression analyses adjusted for sex, smoking, C-reactive
             protein, and current health conditions.<h4>Results</h4>Of
             997 still-living participants, 875 (88%) had plasma suPAR
             measured at age 45. Elevated suPAR was associated with
             accelerated pace of biological aging across multiple organ
             systems, older facial appearance, and with structural signs
             of older brain age. Moreover, participants with higher suPAR
             levels had greater decline in physical function and
             cognitive function from childhood to adulthood compared to
             those with lower suPAR levels. Finally, improvements in
             health habits between ages 38 and 45 (smoking cessation or
             increased physical activity) were associated with less steep
             increases in suPAR levels over those years.<h4>Conclusions</h4>Our
             findings provide initial support for the utility of suPAR in
             studying the role of chronic inflammation in accelerated
             aging and functional decline.},
   Doi = {10.1093/gerona/glaa178},
   Key = {fds364970}
}

@article{fds354222,
   Author = {Gianaros, PJ and Kraynak, TE and Kuan, DC-H and Gross, JJ and McRae, K and Hariri, AR and Manuck, SB and Rasero, J and Verstynen,
             TD},
   Title = {Affective brain patterns as multivariate neural correlates
             of cardiovascular disease risk.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {15},
   Number = {10},
   Pages = {1034-1045},
   Year = {2020},
   Month = {November},
   Abstract = {This study tested whether brain activity patterns evoked by
             affective stimuli relate to individual differences in an
             indicator of pre-clinical atherosclerosis: carotid artery
             intima-media thickness (CA-IMT). Adults (aged 30-54 years)
             completed functional magnetic resonance imaging (fMRI) tasks
             that involved viewing three sets of affective stimuli. Two
             sets included facial expressions of emotion, and one set
             included neutral and unpleasant images from the
             International Affective Picture System (IAPS).
             Cross-validated, multivariate and machine learning models
             showed that individual differences in CA-IMT were partially
             predicted by brain activity patterns evoked by unpleasant
             IAPS images, even after accounting for age, sex and known
             cardiovascular disease risk factors. CA-IMT was also
             predicted by brain activity patterns evoked by angry and
             fearful faces from one of the two stimulus sets of facial
             expressions, but this predictive association did not persist
             after accounting for known cardiovascular risk factors. The
             reliability (internal consistency) of brain activity
             patterns evoked by affective stimuli may have constrained
             their prediction of CA-IMT. Distributed brain activity
             patterns could comprise affective neural correlates of
             pre-clinical atherosclerosis; however, the interpretation of
             such correlates may depend on their psychometric properties,
             as well as the influence of other cardiovascular risk
             factors and specific affective cues.},
   Doi = {10.1093/scan/nsaa050},
   Key = {fds354222}
}

@article{fds364139,
   Author = {Reuben, A and Elliott, ML and Abraham, WC and Broadbent, J and Houts,
             RM and Ireland, D and Knodt, AR and Poulton, R and Ramrakha, S and Hariri,
             AR and Caspi, A and Moffitt, TE},
   Title = {Association of Childhood Lead Exposure With MRI Measurements
             of Structural Brain Integrity in Midlife.},
   Journal = {JAMA},
   Volume = {324},
   Number = {19},
   Pages = {1970-1979},
   Year = {2020},
   Month = {November},
   Abstract = {<h4>Importance</h4>Childhood lead exposure has been linked
             to disrupted brain development, but long-term consequences
             for structural brain integrity are unknown.<h4>Objective</h4>To
             test the hypothesis that childhood lead exposure is
             associated with magnetic resonance imaging (MRI)
             measurements of lower structural integrity of the brain in
             midlife.<h4>Design, setting, and participants</h4>The
             Dunedin Study followed a population-representative 1972-1973
             birth cohort in New Zealand (N = 564 analytic sample) to
             age 45 years (until April 2019).<h4>Exposures</h4>Childhood
             blood lead levels measured at age 11 years.<h4>Main outcomes
             and measures</h4>Structural brain integrity at age 45 years
             assessed via MRI (primary outcomes): gray matter (cortical
             thickness, surface area, hippocampal volume), white matter
             (white matter hyperintensities, fractional anisotropy
             [theoretical range, 0 {diffusion is perfectly isotropic} to
             100 {diffusion is perfectly anisotropic}]), and the Brain
             Age Gap Estimation (BrainAGE), a composite index of the gap
             between chronological age and a machine learning
             algorithm-estimated brain age (0 indicates a brain age
             equivalent to chronological age; positive and negative
             values represent an older and younger brain age,
             respectively). Cognitive function at age 45 years was
             assessed objectively via the Wechsler Adult Intelligence
             Scale IV (IQ range, 40-160, standardized to a mean of 100
             [SD, 15]) and subjectively via informant and self-reports
             (z-score units; scale mean, 0 [SD, 1]).<h4>Results</h4>Of
             1037 original participants, 997 were alive at age 45 years,
             of whom 564 (57%) had received lead testing at age 11 years
             (302 [54%] male) (median follow-up, 34 [interquartile range,
             33.7-34.7] years). Mean blood lead level at age 11 years was
             10.99 (SD, 4.63) μg/dL. After adjusting for covariates,
             each 5-μg/dL higher childhood blood lead level was
             significantly associated with 1.19-cm2 smaller cortical
             surface area (95% CI, -2.35 to -0.02 cm2; P = .05),
             0.10-cm3 smaller hippocampal volume (95% CI, -0.17 to -0.03
             cm3; P = .006), lower global fractional anisotropy
             (b = -0.12; 95% CI, -0.24 to -0.01; P = .04), and a
             BrainAGE index 0.77 years older (95% CI, 0.02-1.51 years;
             P = .05) at age 45 years. There were no statistically
             significant associations between blood lead level and
             log-transformed white matter hyperintensity volume
             (b = 0.05 log mm3; 95% CI, -0.02 to 0.13 log mm3;
             P = .17) or mean cortical thickness (b = -0.004 mm;
             95% CI, -0.012 to 0.004 mm; P = .39). Each 5-μg/dL
             higher childhood blood lead level was significantly
             associated with a 2.07-point lower IQ score at age 45 years
             (95% CI, -3.39 to -0.74; P = .002) and a 0.12-point
             higher score on informant-rated cognitive problems (95% CI,
             0.01-0.23; P = .03). There was no statistically
             significant association between childhood blood lead levels
             and self-reported cognitive problems (b = -0.02 points;
             95% CI, -0.10 to 0.07; P = .68).<h4>Conclusions and
             relevance</h4>In this longitudinal cohort study with a
             median 34-year follow-up, higher childhood blood lead level
             was associated with differences in some MRI measures of
             brain structure that suggested lower structural brain
             integrity in midlife. Because of the large number of
             statistical comparisons, some findings may represent type I
             error.},
   Doi = {10.1001/jama.2020.19998},
   Key = {fds364139}
}

@article{fds364218,
   Author = {Mareckova, K and Hawco, C and Dos Santos and FC and Bakht, A and Calarco,
             N and Miles, AE and Voineskos, AN and Sibille, E and Hariri, AR and Nikolova, YS},
   Title = {Novel polygenic risk score as a translational tool linking
             depression-related changes in the corticolimbic
             transcriptome with neural face processing and anhedonic
             symptoms.},
   Journal = {Translational psychiatry},
   Volume = {10},
   Number = {1},
   Pages = {410},
   Year = {2020},
   Month = {November},
   Abstract = {Convergent data from imaging and postmortem brain
             transcriptome studies implicate corticolimbic circuit (CLC)
             dysregulation in the pathophysiology of depression. To more
             directly bridge these lines of work, we generated a novel
             transcriptome-based polygenic risk score (T-PRS), capturing
             subtle shifts toward depression-like gene expression
             patterns in key CLC regions, and mapped this T-PRS onto
             brain function and related depressive symptoms in a
             nonclinical sample of 478 young adults (225 men; age
             19.79 +/- 1.24) from the Duke Neurogenetics Study.
             First, T-PRS was generated based on common functional SNPs
             shifting CLC gene expression toward a depression-like state.
             Next, we used multivariate partial least squares regression
             to map T-PRS onto whole-brain activity patterns during
             perceptual processing of social stimuli (i.e., human faces).
             For validation, we conducted a comparative analysis with a
             PRS summarizing depression risk variants identified by the
             Psychiatric Genomics Consortium (PGC-PRS). Sex was modeled
             as moderating factor. We showed that T-PRS was associated
             with widespread reductions in neural response to neutral
             faces in women and to emotional faces and shapes in men
             (multivariate p < 0.01). This female-specific reductions
             in neural response to neutral faces was also associated with
             PGC-PRS (multivariate p < 0.03). Reduced reactivity to
             neutral faces was further associated with increased
             self-reported anhedonia. We conclude that women with
             functional alleles mimicking the postmortem transcriptomic
             CLC signature of depression have blunted neural activity to
             social stimuli, which may be expressed as higher
             anhedonia.},
   Doi = {10.1038/s41398-020-01093-w},
   Key = {fds364218}
}

@article{fds364140,
   Author = {d'Arbeloff, T and Cooke, M and Knodt, AR and Sison, M and Melzer, TR and Ireland, D and Poulton, R and Ramrakha, S and Moffitt, TE and Caspi, A and Hariri, AR},
   Title = {Is cardiovascular fitness associated with structural brain
             integrity in midlife? Evidence from a population-representative
             birth cohort study.},
   Journal = {Aging},
   Volume = {12},
   Number = {20},
   Pages = {20888-20914},
   Year = {2020},
   Month = {October},
   Abstract = {Improving cardiovascular fitness may buffer against
             age-related cognitive decline and mitigate dementia risk by
             staving off brain atrophy. However, it is unclear if such
             effects reflect factors operating in childhood
             (neuroselection) or adulthood (neuroprotection). Using data
             from 807 members of the Dunedin Study, a
             population-representative birth cohort, we investigated
             associations between cardiovascular fitness and structural
             brain integrity at age 45, and the extent to which
             associations reflected possible neuroselection or
             neuroprotection by controlling for childhood IQ. Higher
             fitness, as indexed by VO<sub>2</sub>Max, was not associated
             with average cortical thickness, total surface area, or
             subcortical gray matter volume including the hippocampus.
             However, higher fitness was associated with thicker cortex
             in prefrontal and temporal regions as well as greater
             cerebellar gray matter volume. Higher fitness was also
             associated with decreased hippocampal fissure volume. These
             associations were unaffected by the inclusion of childhood
             IQ in analyses. In contrast, a higher rate of decline in
             cardiovascular fitness from 26 to 45 years was not robustly
             associated with structural brain integrity. Our findings are
             consistent with a neuroprotective account of adult
             cardiovascular fitness but suggest that effects are not
             uniformly observed across the brain and reflect
             contemporaneous fitness more so than decline over
             time.},
   Doi = {10.18632/aging.104112},
   Key = {fds364140}
}

@article{fds364219,
   Author = {Avinun, R and Israel, S and Knodt, AR and Hariri,
             AR},
   Title = {Little evidence for associations between the Big Five
             personality traits and variability in brain gray or white
             matter.},
   Journal = {NeuroImage},
   Volume = {220},
   Pages = {117092},
   Year = {2020},
   Month = {October},
   Abstract = {Attempts to link the Big Five personality traits of
             Openness-to-Experience, Conscientiousness, Extraversion,
             Agreeableness, and Neuroticism with variability in
             trait-like features of brain structure have produced
             inconsistent results. Small sample sizes and heterogeneous
             methodology have been suspected in driving these
             inconsistencies. Here, using data collected from 1,107
             university students (636 women, mean age
             19.69 ​± ​1.24 years), representing the largest
             sample to date of unrelated individuals, we tested for
             associations between the Big Five personality traits and
             measures of cortical thickness and surface area, subcortical
             volume, and white matter microstructural integrity. In
             addition to replication analyses based on a prior study, we
             conducted exploratory whole-brain analyses. Four
             supplementary analyses were also conducted to examine 1)
             possible associations with lower-order facets of
             personality; 2) modulatory effects of sex; 3) effect of
             controlling for non-target personality traits; and 4)
             parcellation scheme effects. Our analyses failed to identify
             significant associations between the Big Five personality
             traits and brain morphometry, except for a weak association
             between greater surface area of the superior temporal gyrus
             and lower conscientiousness scores. As the latter
             association is not supported by previous studies, it should
             be treated with caution. Our supplementary analyses mirrored
             these predominantly null findings, suggesting they were not
             substantively biased by our analytic choices. Collectively,
             these results indicate that if there are associations
             between the Big Five personality traits and brain structure,
             they are likely of very small effect size and will require
             very large samples for reliable detection.},
   Doi = {10.1016/j.neuroimage.2020.117092},
   Key = {fds364219}
}

@article{fds350143,
   Author = {Elliott, ML and Knodt, AR and Ireland, D and Morris, ML and Poulton, R and Ramrakha, S and Sison, ML and Moffitt, TE and Caspi, A and Hariri,
             AR},
   Title = {What Is the Test-Retest Reliability of Common
             Task-Functional MRI Measures? New Empirical Evidence and a
             Meta-Analysis.},
   Journal = {Psychological science},
   Volume = {31},
   Number = {7},
   Pages = {792-806},
   Year = {2020},
   Month = {July},
   Abstract = {Identifying brain biomarkers of disease risk is a growing
             priority in neuroscience. The ability to identify meaningful
             biomarkers is limited by measurement reliability; unreliable
             measures are unsuitable for predicting clinical outcomes.
             Measuring brain activity using task functional MRI (fMRI) is
             a major focus of biomarker development; however, the
             reliability of task fMRI has not been systematically
             evaluated. We present converging evidence demonstrating poor
             reliability of task-fMRI measures. First, a meta-analysis of
             90 experiments (<i>N</i> = 1,008) revealed poor overall
             reliability-mean intraclass correlation coefficient (ICC) =
             .397. Second, the test-retest reliabilities of activity in a
             priori regions of interest across 11 common fMRI tasks
             collected by the Human Connectome Project (<i>N</i> = 45)
             and the Dunedin Study (<i>N</i> = 20) were poor (ICCs =
             .067-.485). Collectively, these findings demonstrate that
             common task-fMRI measures are not currently suitable for
             brain biomarker discovery or for individual-differences
             research. We review how this state of affairs came to be and
             highlight avenues for improving task-fMRI
             reliability.},
   Doi = {10.1177/0956797620916786},
   Key = {fds350143}
}

@article{fds364220,
   Author = {Baranger, DAA and Few, LR and Sheinbein, DH and Agrawal, A and Oltmanns,
             TF and Knodt, AR and Barch, DM and Hariri, AR and Bogdan,
             R},
   Title = {Borderline Personality Traits Are Not Correlated With Brain
             Structure in Two Large Samples.},
   Journal = {Biological psychiatry. Cognitive neuroscience and
             neuroimaging},
   Volume = {5},
   Number = {7},
   Pages = {669-677},
   Year = {2020},
   Month = {July},
   Abstract = {<h4>Background</h4>Borderline personality disorder is
             associated with severe psychiatric presentations and has
             been linked to variability in brain structure. Dimensional
             models of borderline personality traits (BPTs) have become
             influential; however, associations between BPTs and brain
             structure remain poorly understood.<h4>Methods</h4>We tested
             whether BPTs are associated with regional cortical
             thickness, cortical surface area, and subcortical volumes
             (n = 152 brain structure metrics) in data from the Duke
             Neurogenetics Study (n = 1299) and Human Connectome Project
             (n = 1099). Positive control analyses tested whether BPTs
             are associated with related behaviors (e.g., suicidal
             thoughts and behaviors, psychiatric diagnoses) and
             experiences (e.g., adverse childhood experiences).<h4>Results</h4>While
             BPTs were robustly associated with all positive control
             measures, they were not significantly associated with any
             brain structure metrics in the Duke Neurogenetics Study or
             Human Connectome Project, or in a meta-analysis of both
             samples. The strongest findings from the meta-analysis
             showed a positive association between BPTs and volumes of
             the left ventral diencephalon and thalamus (p values < .005
             uncorrected, p values > .1 false discovery rate-corrected).
             Contrasting high and low BPT decile groups (n = 552)
             revealed no false discovery rate-significant associations
             with brain structure.<h4>Conclusions</h4>We find replicable
             evidence that BPTs are not associated with brain structure
             despite being correlated with independent behavioral
             measures. Prior reports linking brain morphology to
             borderline personality disorder may be driven by factors
             other than traits (e.g., severe presentations, comorbid
             conditions, severe childhood adversity, or medication) or
             reflect false positives. The etiology or consequences of
             BPTs may not be attributable to brain structure measured via
             magnetic resonance imaging. Future studies of BPTs will
             require much larger sample sizes to detect these very small
             effects.},
   Doi = {10.1016/j.bpsc.2020.02.006},
   Key = {fds364220}
}

@article{fds343722,
   Author = {Puetz, VB and Viding, E and Gerin, MI and Pingault, J-B and Sethi, A and Knodt, AR and Radtke, SR and Brigidi, BD and Hariri, AR and McCrory,
             E},
   Title = {Investigating patterns of neural response associated with
             childhood abuse v. childhood neglect - Corrigendum.},
   Journal = {Psychological medicine},
   Volume = {50},
   Number = {8},
   Pages = {1408},
   Year = {2020},
   Month = {June},
   Abstract = {<h4>Background</h4>Childhood maltreatment is robustly
             associated with increased risk of poor mental health outcome
             and changes in brain function. The authors investigated
             whether childhood experience of abuse (e.g. physical,
             emotional and sexual abuse) and neglect (physical and
             emotional deprivation) was differentially associated with
             neural reactivity to threat.<h4>Methods</h4>Participants
             were drawn from an existing study and allocated to one of
             four groups based on self-report of childhood maltreatment
             experience: individuals with childhood abuse experiences (n
             = 70); individuals with childhood neglect experiences (n =
             87); individuals with combined experience of childhood abuse
             and neglect (n = 50); and non-maltreated individuals (n =
             207) propensity score matched (PSM) on gender, age, IQ,
             psychopathology and SES. Neural reactivity to facial cues
             signalling threat was compared across groups, allowing the
             differential effects associated with particular forms of
             maltreatment experience to be isolated.<h4>Results</h4>Brain
             imaging analyses indicated that while childhood abuse was
             associated with heightened localised threat reactivity in
             ventral amygdala, experiences of neglect were associated
             with heightened reactivity in a distributed cortical
             fronto-parietal network supporting complex social and
             cognitive processing as well as in the dorsal amygdala.
             Unexpectedly, combined experiences of abuse and neglect were
             associated with hypo-activation in several higher-order
             cortical regions as well as the amygdala.<h4>Conclusions</h4>Different
             forms of childhood maltreatment exert differential effects
             in neural threat reactivity: while the effects of abuse are
             more focal, the effects of neglect and combined experiences
             of abuse are more distributed. These findings are relevant
             for understanding the range of psychiatric outcomes
             following childhood maltreatment and have implications for
             intervention.},
   Doi = {10.1017/s0033291719001752},
   Key = {fds343722}
}

@article{fds347193,
   Author = {Baranger, DAA and Demers, CH and Elsayed, NM and Knodt, AR and Radtke,
             SR and Desmarais, A and Few, LR and Agrawal, A and Heath, AC and Barch, DM and Squeglia, LM and Williamson, DE and Hariri, AR and Bogdan,
             R},
   Title = {Convergent Evidence for Predispositional Effects of Brain
             Gray Matter Volume on Alcohol Consumption.},
   Journal = {Biological psychiatry},
   Volume = {87},
   Number = {7},
   Pages = {645-655},
   Year = {2020},
   Month = {April},
   Abstract = {<h4>Background</h4>Alcohol use has been reliably associated
             with smaller subcortical and cortical regional gray matter
             volumes (GMVs). Whether these associations reflect shared
             predisposing risk factors or causal consequences of alcohol
             use remains poorly understood.<h4>Methods</h4>Data came from
             3 neuroimaging samples (N = 2423), spanning childhood or
             adolescence to middle age, with prospective or family-based
             data. First, we identified replicable GMV correlates of
             alcohol use. Next, we used family-based and longitudinal
             data to test whether these associations may plausibly
             reflect a predispositional liability for alcohol use or a
             causal consequence of alcohol use. Finally, we used
             heritability, gene-set enrichment, and transcriptome-wide
             association study approaches to evaluate whether genome-wide
             association study-defined genomic risk for alcohol
             consumption is enriched for genes that are preferentially
             expressed in regions that were identified in our
             neuroimaging analyses.<h4>Results</h4>Smaller right
             dorsolateral prefrontal cortex (DLPFC) (i.e., middle and
             superior frontal gyri) and insula GMVs were associated with
             increased alcohol use across samples. Family-based and
             prospective longitudinal data suggest that these
             associations are genetically conferred and that DLPFC GMV
             prospectively predicts future use and initiation. Genomic
             risk for alcohol use was enriched in gene sets that were
             preferentially expressed in the DLPFC and was associated
             with replicable differential gene expression in the
             DLPFC.<h4>Conclusions</h4>These data suggest that smaller
             DLPFC and insula GMV plausibly represent genetically
             conferred predispositional risk factors for, as opposed to
             consequences of, alcohol use. DLPFC and insula GMV represent
             promising biomarkers for alcohol-consumption liability and
             related psychiatric and behavioral phenotypes.},
   Doi = {10.1016/j.biopsych.2019.08.029},
   Key = {fds347193}
}

@article{fds364147,
   Author = {Wallace, GL and Richard, E and Peng, CS and Knodt, AR and Hariri,
             AR},
   Title = {Subclinical eating disorder traits are correlated with
             cortical thickness in regions associated with food reward
             and perception.},
   Journal = {Brain imaging and behavior},
   Volume = {14},
   Number = {2},
   Pages = {346-352},
   Year = {2020},
   Month = {April},
   Abstract = {Behavioral traits associated with various forms of
             psychopathology are conceptualized as dimensional, varying
             from those present in a frank disorder to subclinical
             expression. Demonstrating links between these behavioral
             traits and neurobiological indicators, such as brain
             structure, provides one form of validation for this view.
             However, unlike behavioral dimensions associated with other
             forms of psychopathology (e.g., autism spectrum disorder,
             attention deficit hyperactivity disorder, antisocial
             disorders), eating disorder traits have not been
             investigated in this manner in spite of the potential that
             such an approach has to elucidate etiological mechanisms.
             Therefore, we examined for the first time neural
             endophenotypes of Anorexia Nervosa and Bulimia via
             dimensional traits (measured using the Eating Disorders
             Inventory-3) in a large subclinical sample of young adults
             (n = 456 and n = 247, respectively;
             ages = 18-22 years) who each provided a structural
             magnetic resonance imaging scan. Cortical thickness was
             quantified at 81,924 vertices across the cortical surface.
             We found: 1) increasing eating disorder traits correlated
             with thinner cortex in the insula and orbitofrontal cortex,
             among other regions, and 2) using these regions as seeds,
             increasing eating disorder trait scores negatively modulated
             structural covariance between these seed regions and other
             cortical regions linked to regulatory and sensorimotor
             functions (e.g., frontal and temporal cortices). These
             findings parallel those found in the clinical literature
             (i.e., thinner cortex in these food-related regions in
             individuals with eating disorders) and therefore provide
             evidence supporting the dimensional view of behavioral
             traits associated with eating disorders. Extending this
             approach to genetic and neuroimaging genetics studies holds
             promise to inform etiology.},
   Doi = {10.1007/s11682-018-0007-x},
   Key = {fds364147}
}

@article{fds364971,
   Author = {Caspi, A and Houts, RM and Ambler, A and Danese, A and Elliott, ML and Hariri, A and Harrington, H and Hogan, S and Poulton, R and Ramrakha, S and Rasmussen, LJH and Reuben, A and Richmond-Rakerd, L and Sugden, K and Wertz, J and Williams, BS and Moffitt, TE},
   Title = {Longitudinal Assessment of Mental Health Disorders and
             Comorbidities Across 4 Decades Among Participants in the
             Dunedin Birth Cohort Study.},
   Journal = {JAMA network open},
   Volume = {3},
   Number = {4},
   Pages = {e203221},
   Year = {2020},
   Month = {April},
   Abstract = {<h4>Importance</h4>Mental health professionals typically
             encounter patients at 1 point in patients' lives. This
             cross-sectional window understandably fosters focus on the
             current presenting diagnosis. Research programs, treatment
             protocols, specialist clinics, and specialist journals are
             oriented to presenting diagnoses, on the assumption that
             diagnosis informs about causes and prognosis. This study
             tests an alternative hypothesis: people with mental
             disorders experience many different kinds of disorders
             across diagnostic families, when followed for 4
             decades.<h4>Objective</h4>To describe mental disorder life
             histories across the first half of the life
             course.<h4>Design, setting, and participants</h4>This cohort
             study involved participants born in New Zealand from 1972 to
             1973 who were enrolled in the population-representative
             Dunedin Study. Participants were observed from birth to age
             45 years (until April 2019). Data were analyzed from May
             2019 to January 2020.<h4>Main outcomes and
             measures</h4>Diagnosed impairing disorders were assessed 9
             times from ages 11 to 45 years. Brain function was assessed
             through neurocognitive examinations conducted at age 3
             years, neuropsychological testing during childhood and
             adulthood, and midlife neuroimaging-based brain
             age.<h4>Results</h4>Of 1037 original participants (535 male
             [51.6%]), 1013 had mental health data available. The
             proportions of participants meeting the criteria for a
             mental disorder were as follows: 35% (346 of 975) at ages 11
             to 15 years, 50% (473 of 941) at age 18 years, 51% (489 of
             961) at age 21 years, 48% (472 of 977) at age 26 years, 46%
             (444 of 969) at age 32 years, 45% (429 of 955) at age 38
             years, and 44% (407 of 927) at age 45 years. The onset of
             the disorder occurred by adolescence for 59% of participants
             (600 of 1013), eventually affecting 86% of the cohort (869
             of 1013) by midlife. By age 45 years, 85% of participants
             (737 of 869) with a disorder had accumulated comorbid
             diagnoses. Participants with adolescent-onset disorders
             subsequently presented with disorders at more past-year
             assessments (r = 0.71; 95% CI, 0.68 to 0.74;
             P < .001) and met the criteria for more diverse
             disorders (r = 0.64; 95% CI, 0.60 to 0.67;
             P < .001). Confirmatory factor analysis summarizing
             mental disorder life histories across 4 decades identified a
             general factor of psychopathology, the p-factor.
             Longitudinal analyses showed that high p-factor scores
             (indicating extensive mental disorder life histories) were
             antedated by poor neurocognitive functioning at age 3 years
             (r = -0.18; 95% CI, -0.24 to -0.12; P < .001), were
             accompanied by childhood-to-adulthood cognitive decline
             (r = -0.11; 95% CI, -0.17 to -0.04; P < .001), and
             were associated with older brain age at midlife
             (r = 0.14; 95% CI, 0.07 to 0.20; P < .001).<h4>Conclusions
             and relevance</h4>These findings suggest that mental
             disorder life histories shift among different successive
             disorders. Data from the present study, alongside nationwide
             data from Danish health registers, inform a life-course
             perspective on mental disorders. This perspective cautions
             against overreliance on diagnosis-specific research and
             clinical protocols.},
   Doi = {10.1001/jamanetworkopen.2020.3221},
   Key = {fds364971}
}

@article{fds344832,
   Author = {Avinun, R and Nevo, A and Radtke, SR and Brigidi, BD and Hariri,
             AR},
   Title = {Divergence of an association between depressive symptoms and
             a dopamine polygenic score in Caucasians and
             Asians.},
   Journal = {European archives of psychiatry and clinical
             neuroscience},
   Volume = {270},
   Number = {2},
   Pages = {229-235},
   Year = {2020},
   Month = {March},
   Abstract = {A recent study reported a negative association between a
             putatively functional dopamine (DA) polygenic score,
             indexing higher levels of DA signaling, and depressive
             symptoms. We attempted to replicate this association using
             data from the Duke Neurogenetics Study. Our replication
             attempt was made in a subsample of 520 non-Hispanic
             Caucasian volunteers (277 women, mean age
             19.78 ± 1.24 years). The DA polygenic score was based
             on the following five loci: rs27072 (SLC6A3/DAT1), rs4532
             (DRD1), rs1800497 (DRD2/ANKK1), rs6280 (DRD3), and rs4680
             (COMT). Because the discovery sample in the original study
             consisted mostly of Asian participants, we also conducted a
             post hoc analysis in a smaller subsample of Asian volunteers
             (N = 316, 179 women, mean age 19.61 ± 1.32 years).
             In the primary sample of non-Hispanic Caucasians, a linear
             regression analysis controlling for sex, age, socioeconomic
             status (SES), body mass index, genetic ancestry, and both
             early and recent life stress, revealed that higher DA
             polygenic scores were associated with higher self-reported
             symptoms of depression. This was in contrast to the original
             association of higher DA polygenic scores and lower
             depressive symptoms. However, the direction of the
             association in our Asian subsample was consistent with this
             original finding. Our results also suggested that compared
             to the Asian subsample, the non-Hispanic Caucasian subsample
             was characterized by higher SES, lower early and recent life
             stress, and lower depressive symptoms. These differences may
             have contributed to the observed divergence in associations.
             Collectively, the current findings add to evidence that
             specific genetic associations may differ between populations
             and further encourage explicit modeling of race/ethnicity in
             examining the polygenic nature of depressive symptoms and
             depression.},
   Doi = {10.1007/s00406-019-01040-x},
   Key = {fds344832}
}

@article{fds364972,
   Author = {Carlisi, CO and Moffitt, TE and Knodt, AR and Harrington, H and Ireland,
             D and Melzer, TR and Poulton, R and Ramrakha, S and Caspi, A and Hariri,
             AR and Viding, E},
   Title = {Associations between life-course-persistent antisocial
             behaviour and brain structure in a population-representative
             longitudinal birth cohort.},
   Journal = {The lancet. Psychiatry},
   Volume = {7},
   Number = {3},
   Pages = {245-253},
   Year = {2020},
   Month = {March},
   Abstract = {<h4>Background</h4>Studies with behavioural and
             neuropsychological tests have supported the developmental
             taxonomy theory of antisocial behaviour, which specifies
             abnormal brain development as a fundamental aspect of
             life-course-persistent antisocial behaviour, but no study
             has characterised features of brain structure associated
             with life-course-persistent versus adolescence-limited
             trajectories, as defined by prospective data. We aimed to
             determine whether life-course-persistent antisocial
             behaviour is associated with neurocognitive abnormalities by
             testing the hypothesis that it is also associated with brain
             structure abnormalities.<h4>Methods</h4>We used structural
             MRI data collected at 45 years of age from participants in
             the Dunedin Study, a population-representative longitudinal
             birth cohort of 1037 individuals born between April 1, 1972,
             and March 31, 1973, in Dunedin, New Zealand, who were
             resident in the province and who participated in the first
             assessment at 3 years of age. Participants underwent MRI,
             and mean global cortical surface area and cortical thickness
             were extracted for each participant. Participants had been
             previously subtyped as exhibiting life-course-persistent,
             adolescence-limited, or no history of persistent antisocial
             behaviour (ie, a low trajectory group) based on
             informant-reported and self-reported conduct problems from
             the ages of 7 years to 26 years. Study personnel who
             processed the MRI images were masked to antisocial group
             membership. We used linear estimated ordinary least squares
             regressions to compare each antisocial trajectory group
             (life-course persistent and adolescence limited) with the
             low trajectory group to examine whether antisocial behaviour
             was related to abnormalities in mean global surface area and
             mean cortical thickness. Next, we used parcel-wise linear
             regressions to identify antisocial trajectory group
             differences in surface area and cortical thickness. All
             results were controlled for sex and false discovery rate
             corrected.<h4>Findings</h4>Data from 672 participants were
             analysed, and 80 (12%) were classified as having
             life-course-persistent antisocial behaviour, 151 (23%) as
             having adolescence-limited antisocial behaviour, and 441
             (66%) as having low antisocial behaviour. Individuals on the
             life-course-persistent trajectory had a smaller mean surface
             area (standardised β=-0·18 [95% CI -0·24 to -0·11];
             p<0·0001) and lower mean cortical thickness (standardised
             β=-0·10 [95% CI -0·19 to -0·02]; p=0·020) than did
             those in the low group. Compared with the low group, the
             life-course-persistent group had reduced surface area in 282
             of 360 anatomically defined parcels and thinner cortex in 11
             of 360 parcels encompassing circumscribed frontal and
             temporal regions associated with executive function, affect
             regulation, and motivation. Widespread differences in brain
             surface morphometry were not observed for the
             adolescence-limited group compared with either
             non-antisocial behaviour or life-course-persistent
             groups.<h4>Interpretation</h4>These analyses provide initial
             evidence that differences in brain surface morphometry are
             associated with life-course-persistent, but not
             adolescence-limited, antisocial behaviour. As such, the
             analyses are consistent with the developmental taxonomy
             theory of antisocial behaviour and highlight the importance
             of using prospective longitudinal data to define different
             patterns of antisocial behaviour development.<h4>Funding</h4>US
             National Institute on Aging, Health Research Council of New
             Zealand, New Zealand Ministry of Business, Innovation and
             Employment, UK Medical Research Council, Avielle Foundation,
             and Wellcome Trust.},
   Doi = {10.1016/s2215-0366(20)30002-x},
   Key = {fds364972}
}

@article{fds364172,
   Author = {Burr, DA and d'Arbeloff, T and Elliott, ML and Knodt, AR and Brigidi,
             BD and Hariri, AR},
   Title = {Functional connectivity predicts the dispositional use of
             expressive suppression but not cognitive
             reappraisal.},
   Journal = {Brain and behavior},
   Volume = {10},
   Number = {2},
   Pages = {e01493},
   Year = {2020},
   Month = {February},
   Abstract = {<h4>Introduction</h4>Previous research has identified
             specific brain regions associated with regulating emotion
             using common strategies such as expressive suppression and
             cognitive reappraisal. However, most research focuses on a
             priori regions and directs participants how to regulate,
             which may not reflect how people naturally regulate outside
             the laboratory.<h4>Method</h4>Here, we used a data-driven
             approach to investigate how individual differences in
             distributed intrinsic functional brain connectivity predict
             emotion regulation tendency outside the laboratory.
             Specifically, we used connectome-based predictive modeling
             to extract functional connections in the brain significantly
             related to the dispositional use of suppression and
             reappraisal. These edges were then used in a predictive
             model and cross-validated in novel participants to identify
             a neural signature that reflects individual differences in
             the tendency to suppress and reappraise emotion.<h4>Results</h4>We
             found a significant neural signature for the dispositional
             use of suppression, but not reappraisal. Within this
             whole-brain signature, the intrinsic connectivity of the
             default mode network was most informative of suppression
             tendency. In addition, the predictive performance of this
             model was significant in males, but not females.<h4>Conclusion</h4>These
             findings help inform how whole-brain networks of functional
             connectivity characterize how people tend to regulate
             emotion outside the laboratory.},
   Doi = {10.1002/brb3.1493},
   Key = {fds364172}
}

@article{fds364142,
   Author = {Detloff, AM and Hariri, AR and Strauman, TJ},
   Title = {Neural signatures of promotion versus prevention goal
             priming: fMRI evidence for distinct cognitive-motivational
             systems.},
   Journal = {Personality neuroscience},
   Volume = {3},
   Pages = {e1},
   Year = {2020},
   Month = {February},
   Abstract = {Regulatory focus theory (RFT) postulates two
             cognitive-motivational systems for personal goal pursuit:
             the promotion system, which is associated with ideal goals
             (an individual's hopes, dreams, and aspirations), and the
             prevention system, which is associated with ought goals (an
             individual's duties, responsibilities, and obligations). The
             two systems have been studied extensively in behavioral
             research with reference to differences between promotion and
             prevention goal pursuit as well as the consequences of
             perceived attainment versus nonattainment within each
             system. However, no study has examined the neural correlates
             of each combination of goal domain and goal attainment
             status. We used a rapid masked idiographic goal priming
             paradigm and functional magnetic resonance imaging to
             present individually selected promotion and prevention
             goals, which participants had reported previously that they
             were close to attaining ("match") or far from attaining
             ("mismatch"). Across the four priming conditions,
             significant activations were observed in bilateral insula
             (Brodmann area (BA) 13) and visual association cortex (BA
             18/19). Promotion priming discriminantly engaged left
             prefrontal cortex (BA 9), whereas prevention priming
             discriminantly engaged right prefrontal cortex (BA 8/9).
             Activation in response to promotion goal priming was also
             correlated with an individual difference measure of
             perceived success in promotion goal attainment. Our findings
             extend the construct validity of RFT by showing that the two
             systems postulated by RFT, under conditions of both
             attainment and nonattainment, have shared and distinct
             neural correlates that interface logically with established
             network models of self-regulatory cognition.},
   Doi = {10.1017/pen.2019.13},
   Key = {fds364142}
}

@article{fds364134,
   Author = {Lee, MR and Shin, JH and Deschaine, S and Daurio, AM and Stangl, BL and Yan, J and Ramchandani, VA and Schwandt, ML and Grodin, EN and Momenan,
             R and Corral-Frias, NS and Hariri, AR and Bogdan, R and Alvarez, VA and Leggio, L},
   Title = {A role for the CD38 rs3796863 polymorphism in alcohol
             and monetary reward: evidence from CD38 knockout mice and
             alcohol self-administration, [11C]-raclopride binding, and
             functional MRI in humans.},
   Journal = {The American journal of drug and alcohol
             abuse},
   Volume = {46},
   Number = {2},
   Pages = {167-179},
   Year = {2020},
   Month = {January},
   Abstract = {<i>Background</i>: Cluster of differentiation 38 (CD38) is a
             transmembrane protein expressed in dopaminergic reward
             pathways in the brain, including the nucleus accumbens
             (NAc). The GG genotype of a common single nucleotide
             polymorphism (SNP) within <i>CD38</i>, rs3796863, is
             associated with increased social reward.<i>Objective</i>:
             Examine whether <i>CD38</i> rs3796863 and <i>Cd38</i>
             knockout (KO) are associated with reward-related neural and
             behavioral phenotypes.<i>Methods</i>: Data from four
             independent human studies were used to test whether
             rs3796863 genotype is associated with: (1) intravenous
             alcohol self-administration (n = 64, 30 females), (2)
             alcohol-stimulated dopamine (DA) release measured using
             <sup>11</sup>C-raclopride positron emission tomography
             (n = 22 men), (3) ventral striatum (VS) response to
             positive feedback measured using a card guessing functional
             magnetic resonance imaging (fMRI) paradigm (n = 531, 276
             females), and (4) resting state functional connectivity
             (rsfc) of the VS (n = 51, 26 females). In a fifth study,
             we used a mouse model to examine whether <i>cd38</i>
             knockout influences stimulated DA release in the NAc core
             and dorsal striatum using fast-scanning cyclic
             voltammetry.<i>Results</i>: Relative to T allele carriers, G
             homozygotes at rs3796863 within <i>CD38</i> were
             characterized by greater alcohol self-administration,
             alcohol-stimulated dopamine release, VS response to positive
             feedback, and rsfc between the VS and anterior cingulate
             cortex. High-frequency stimulation reduced DA release among
             <i>Cd38</i> KO mice had reduced dopamine release in the
             NAc.<i>Conclusion</i>: Converging evidence suggests that
             <i>CD38</i> rs3796863 genotype may increase DA-related
             reward response and alcohol consumption.},
   Doi = {10.1080/00952990.2019.1638928},
   Key = {fds364134}
}

@article{fds346711,
   Author = {Farber, MJ and Kim, MJ and Knodt, AR and Hariri, AR},
   Title = {Maternal overprotection in childhood is associated with
             amygdala reactivity and structural connectivity in
             adulthood.},
   Journal = {Developmental cognitive neuroscience},
   Volume = {40},
   Pages = {100711},
   Year = {2019},
   Month = {December},
   Abstract = {Recently, we reported that variability in early-life
             caregiving experiences maps onto individual differences in
             threat-related brain function. Here, we extend this work to
             provide further evidence that subtle variability in specific
             features of early caregiving shapes structural and
             functional connectivity between the amygdala and medial
             prefrontal cortex (mPFC) in a cohort of 312 young adult
             volunteers. Multiple regression analyses revealed that
             participants who reported higher maternal overprotection
             exhibited increased amygdala reactivity to explicit signals
             of interpersonal threat but not implicit signals of broad
             environmental threat. While amygdala functional connectivity
             with regulatory regions of the mPFC was not significantly
             associated with maternal overprotection, participants who
             reported higher maternal overprotection exhibited relatively
             decreased structural integrity of the uncinate fasciculus
             (UF), a white matter tract connecting these same brain
             regions. There were no significant associations between
             structural or functional brain measures and either maternal
             or paternal care ratings. These findings suggest that an
             overprotective maternal parenting style during childhood is
             associated with later functional and structural alterations
             of brain regions involved in generating and regulating
             responses to threat.},
   Doi = {10.1016/j.dcn.2019.100711},
   Key = {fds346711}
}

@article{fds346872,
   Author = {Wetherill, L and Lai, D and Johnson, EC and Anokhin, A and Bauer, L and Bucholz, KK and Dick, DM and Hariri, AR and Hesselbrock, V and Kamarajan, C and Kramer, J and Kuperman, S and Meyers, JL and Nurnberger, JI and Schuckit, M and Scott, DM and Taylor, RE and Tischfield, J and Porjesz, B and Goate, AM and Edenberg, HJ and Foroud,
             T and Bogdan, R and Agrawal, A},
   Title = {ERRATUM: Genome-wide association study identifies loci
             associated with liability to alcohol and drug dependence
             that is associated with variability in reward-related
             ventral striatum activity in African- and
             European-Americans.},
   Journal = {Genes, brain, and behavior},
   Volume = {18},
   Number = {8},
   Pages = {e12608},
   Year = {2019},
   Month = {November},
   Doi = {10.1111/gbb.12608},
   Key = {fds346872}
}

@article{fds364148,
   Author = {Avinun, R and Hariri, AR},
   Title = {A polygenic score for body mass index is associated with
             depressive symptoms via early life stress: Evidence for
             gene-environment correlation.},
   Journal = {Journal of psychiatric research},
   Volume = {118},
   Pages = {9-13},
   Year = {2019},
   Month = {November},
   Abstract = {Increasing childhood obesity rates are associated with not
             only adverse physical, but also mental health outcomes,
             including depression. These negative outcomes may be caused
             and/or exacerbated by the bullying and shaming overweight
             individuals experience. As body mass index (BMI) can be
             highly heritable, we hypothesized that a genetic risk for
             higher BMI, will predict higher early life stress (ELS),
             which in turn will predict higher depressive symptoms in
             adulthood. Such a process will reflect an evocative
             gene-environment correlation (rGE) wherein an individual's
             genetically influenced phenotype evokes a reaction from the
             environment that subsequently shapes the individual's
             health. We modeled genetic risk using a polygenic score of
             BMI derived from a recent large GWAS meta-analysis.
             Self-reports were used for the assessment of ELS and
             depressive symptoms in adulthood. The discovery sample
             consisted of 524 non-Hispanic Caucasian university students
             from the Duke Neurogenetics Study (DNS; 278 women, mean age
             19.78 ± 1.23 years) and the independent replication
             sample consisted of 5930 white British individuals from the
             UK biobank (UKB; 3128 women, mean age 62.66 ± 7.38
             years). A significant mediation effect was found in the DNS
             (indirect effect = 0.207, bootstrapped SE = .10,
             bootstrapped 95% CI: 0.014 to 0.421), and then replicated in
             the UKB (indirect effect = 0.04, bootstrapped
             SE = .01, bootstrapped 95% CI: 0.018 to 0.066). Higher
             BMI polygenic scores predicted higher ELS, which in turn
             predicted higher depressive symptoms. Our findings suggest
             that evocative rGE may contribute to weight-related mental
             health problems and stress the need for interventions that
             aim to reduce weight bias, specifically during
             childhood.},
   Doi = {10.1016/j.jpsychires.2019.08.008},
   Key = {fds364148}
}

@article{fds364973,
   Author = {Rasmussen, LJH and Caspi, A and Ambler, A and Broadbent, JM and Cohen,
             HJ and d'Arbeloff, T and Elliott, M and Hancox, RJ and Harrington, H and Hogan, S and Houts, R and Ireland, D and Knodt, AR and Meredith-Jones,
             K and Morey, MC and Morrison, L and Poulton, R and Ramrakha, S and Richmond-Rakerd, L and Sison, ML and Sneddon, K and Thomson, WM and Hariri, AR and Moffitt, TE},
   Title = {Association of Neurocognitive and Physical Function With
             Gait Speed in Midlife.},
   Journal = {JAMA Netw Open},
   Volume = {2},
   Number = {10},
   Pages = {e1913123},
   Year = {2019},
   Month = {October},
   Abstract = {IMPORTANCE: Gait speed is a well-known indicator of risk of
             functional decline and mortality in older adults, but little
             is known about the factors associated with gait speed
             earlier in life. OBJECTIVES: To test the hypothesis that
             slow gait speed reflects accelerated biological aging at
             midlife, as well as poor neurocognitive functioning in
             childhood and cognitive decline from childhood to midlife.
             DESIGN, SETTING, AND PARTICIPANTS: This cohort study uses
             data from the Dunedin Multidisciplinary Health and
             Development Study, a population-based study of a
             representative 1972 to 1973 birth cohort in New Zealand that
             observed participants to age 45 years (until April 2019).
             Data analysis was performed from April to June 2019.
             EXPOSURES: Childhood neurocognitive functions and
             accelerated aging, brain structure, and concurrent physical
             and cognitive functions in adulthood. MAIN OUTCOMES AND
             MEASURES: Gait speed at age 45 years, measured under 3
             walking conditions: usual, dual task, and maximum gait
             speeds. RESULTS: Of the 1037 original participants (91% of
             eligible births; 535 [51.6%] male), 997 were alive at age 45
             years, of whom 904 (90.7%) had gait speed measured (455
             [50.3%] male; 93% white). The mean (SD) gait speeds were
             1.30 (0.17) m/s for usual gait, 1.16 (0.23) m/s for dual
             task gait, and 1.99 (0.29) m/s for maximum gait. Adults with
             more physical limitations (standardized regression
             coefficient [β], -0.27; 95% CI, -0.34 to -0.21;
             P < .001), poorer physical functions (ie, weak grip
             strength [β, 0.36; 95% CI, 0.25 to 0.46], poor balance [β,
             0.28; 95% CI, 0.21 to 0.34], poor visual-motor coordination
             [β, 0.24; 95% CI, 0.17 to 0.30], and poor performance on
             the chair-stand [β, 0.34; 95% CI, 0.27 to 0.40] or 2-minute
             step tests [β, 0.33; 95% CI, 0.27 to 0.39]; all
             P < .001), accelerated biological aging across multiple
             organ systems (β, -0.33; 95% CI, -0.40 to -0.27;
             P < .001), older facial appearance (β, -0.25; 95% CI,
             -0.31 to -0.18; P < .001), smaller brain volume (β,
             0.15; 95% CI, 0.06 to 0.23; P < .001), more cortical
             thinning (β, 0.09; 95% CI, 0.02 to 0.16; P = .01),
             smaller cortical surface area (β, 0.13; 95% CI, 0.04 to
             0.21; P = .003), and more white matter hyperintensities
             (β, -0.09; 95% CI, -0.15 to -0.02; P = .01) had slower
             gait speed. Participants with lower IQ in midlife (β, 0.38;
             95% CI, 0.32 to 0.44; P < .001) and participants who
             exhibited cognitive decline from childhood to adulthood (β,
             0.10; 95% CI, 0.04 to 0.17; P < .001) had slower gait at
             age 45 years. Those with poor neurocognitive functioning as
             early as age 3 years had slower gait in midlife (β, 0.26;
             95% CI, 0.20 to 0.32; P < .001). CONCLUSIONS AND
             RELEVANCE: Adults' gait speed is associated with more than
             geriatric functional status; it is also associated with
             midlife aging and lifelong brain health.},
   Doi = {10.1001/jamanetworkopen.2019.13123},
   Key = {fds364973}
}

@article{fds364149,
   Author = {Avinun, R and Nevo, A and Knodt, AR and Elliott, ML and Hariri,
             AR},
   Title = {A genome-wide association study-derived polygenic score for
             interleukin-1β is associated with hippocampal volume in two
             samples.},
   Journal = {Human brain mapping},
   Volume = {40},
   Number = {13},
   Pages = {3910-3917},
   Year = {2019},
   Month = {September},
   Abstract = {Accumulating research suggests that the pro-inflammatory
             cytokine interleukin-1β (IL-1β) has a modulatory effect on
             the hippocampus, a brain structure important for learning
             and memory as well as linked with both psychiatric and
             neurodegenerative disorders. Here, we used an imaging
             genetics strategy to test an association between an IL-1β
             polygenic score and hippocampal volume in two independent
             samples. Our polygenic score was derived using summary
             statistics from a recent genome-wide association study of
             circulating cytokines that included IL-1β (N = 3,309). In
             the first sample of 512 non-Hispanic Caucasian university
             students (274 women, mean age 19.78 ± 1.24 years) from
             the Duke Neurogenetics Study, we identified a significant
             positive correlation between IL-1β polygenic scores and
             hippocampal volume. This positive association was
             successfully replicated in a second sample of 7,960 white
             British volunteers (4,158 women, mean age 62.63 ± 7.45
             years) from the UK Biobank. Our results lend further support
             in humans, to the link between IL-1β and the structure of
             the hippocampus.},
   Doi = {10.1002/hbm.24639},
   Key = {fds364149}
}

@article{fds345489,
   Author = {Lam, M and Hill, WD and Trampush, JW and Yu, J and Knowles, E and Davies,
             G and Stahl, E and Huckins, L and Liewald, DC and Djurovic, S and Melle, I and Sundet, K and Christoforou, A and Reinvang, I and DeRosse, P and Lundervold, AJ and Steen, VM and Espeseth, T and Räikkönen, K and Widen, E and Palotie, A and Eriksson, JG and Giegling, I and Konte, B and Hartmann, AM and Roussos, P and Giakoumaki, S and Burdick, KE and Payton, A and Ollier, W and Chiba-Falek, O and Attix, DK and Need, AC and Cirulli, ET and Voineskos, AN and Stefanis, NC and Avramopoulos, D and Hatzimanolis, A and Arking, DE and Smyrnis, N and Bilder, RM and Freimer, NA and Cannon, TD and London, E and Poldrack, RA and Sabb, FW and Congdon, E and Conley, ED and Scult, MA and Dickinson, D and Straub, RE and Donohoe, G and Morris, D and Corvin, A and Gill, M and Hariri, AR and Weinberger, DR and Pendleton, N and Bitsios, P and Rujescu, D and Lahti,
             J and Le Hellard and S and Keller, MC and Andreassen, OA and Deary, IJ and Glahn, DC and Malhotra, AK and Lencz, T},
   Title = {Pleiotropic Meta-Analysis of Cognition, Education, and
             Schizophrenia Differentiates Roles of Early
             Neurodevelopmental and Adult Synaptic Pathways.},
   Journal = {Am J Hum Genet},
   Volume = {105},
   Number = {2},
   Pages = {334-350},
   Year = {2019},
   Month = {August},
   Abstract = {Susceptibility to schizophrenia is inversely correlated with
             general cognitive ability at both the phenotypic and the
             genetic level. Paradoxically, a modest but consistent
             positive genetic correlation has been reported between
             schizophrenia and educational attainment, despite the strong
             positive genetic correlation between cognitive ability and
             educational attainment. Here we leverage published
             genome-wide association studies (GWASs) in cognitive
             ability, education, and schizophrenia to parse biological
             mechanisms underlying these results. Association analysis
             based on subsets (ASSET), a pleiotropic meta-analytic
             technique, allowed jointly associated loci to be identified
             and characterized. Specifically, we identified subsets of
             variants associated in the expected ("concordant") direction
             across all three phenotypes (i.e., greater risk for
             schizophrenia, lower cognitive ability, and lower
             educational attainment); these were contrasted with variants
             that demonstrated the counterintuitive ("discordant")
             relationship between education and schizophrenia (i.e.,
             greater risk for schizophrenia and higher educational
             attainment). ASSET analysis revealed 235 independent loci
             associated with cognitive ability, education, and/or
             schizophrenia at p < 5 × 10-8. Pleiotropic analysis
             successfully identified more than 100 loci that were not
             significant in the input GWASs. Many of these have been
             validated by larger, more recent single-phenotype GWASs.
             Leveraging the joint genetic correlations of cognitive
             ability, education, and schizophrenia, we were able to
             dissociate two distinct biological mechanisms-early
             neurodevelopmental pathways that characterize concordant
             allelic variation and adulthood synaptic pruning
             pathways-that were linked to the paradoxical positive
             genetic association between education and schizophrenia.
             Furthermore, genetic correlation analyses revealed that
             these mechanisms contribute not only to the etiopathogenesis
             of schizophrenia but also to the broader biological
             dimensions implicated in both general health outcomes and
             psychiatric illness.},
   Doi = {10.1016/j.ajhg.2019.06.012},
   Key = {fds345489}
}

@article{fds343721,
   Author = {Lai, D and Wetherill, L and Bertelsen, S and Carey, CE and Kamarajan, C and Kapoor, M and Meyers, JL and Anokhin, AP and Bennett, DA and Bucholz,
             KK and Chang, KK and De Jager and PL and Dick, DM and Hesselbrock, V and Kramer, J and Kuperman, S and Nurnberger, JI and Raj, T and Schuckit, M and Scott, DM and Taylor, RE and Tischfield, J and Hariri, AR and Edenberg,
             HJ and Agrawal, A and Bogdan, R and Porjesz, B and Goate, AM and Foroud,
             T},
   Title = {Genome-wide association studies of alcohol dependence,
             DSM-IV criterion count and individual criteria.},
   Journal = {Genes, brain, and behavior},
   Volume = {18},
   Number = {6},
   Pages = {e12579},
   Year = {2019},
   Month = {July},
   Abstract = {Genome-wide association studies (GWAS) of alcohol dependence
             (AD) have reliably identified variation within alcohol
             metabolizing genes (eg, ADH1B) but have inconsistently
             located other signals, which may be partially attributable
             to symptom heterogeneity underlying the disorder. We
             conducted GWAS of DSM-IV AD (primary analysis), DSM-IV AD
             criterion count (secondary analysis), and individual
             dependence criteria (tertiary analysis) among 7418 (1121
             families) European American (EA) individuals from the
             Collaborative Study on the Genetics of Alcoholism (COGA).
             Trans-ancestral meta-analyses combined these results with
             data from 3175 (585 families) African-American (AA)
             individuals from COGA. In the EA GWAS, three loci were
             genome-wide significant: rs1229984 in ADH1B for AD criterion
             count (P = 4.16E-11) and Desire to cut drinking (P =
             1.21E-11); rs188227250 (chromosome 8, Drinking more than
             intended, P = 6.72E-09); rs1912461 (chromosome 15, Time
             spent drinking, P = 1.77E-08). In the trans-ancestral
             meta-analysis, rs1229984 was associated with multiple
             phenotypes and two additional loci were genome-wide
             significant: rs61826952 (chromosome 1, DSM-IV AD, P =
             8.42E-11); rs7597960 (chromosome 2, Time spent drinking, P =
             1.22E-08). Associations with rs1229984 and rs18822750 were
             replicated in independent datasets. Polygenic risk scores
             derived from the EA GWAS of AD predicted AD in two EA
             datasets (P < .01; 0.61%-1.82% of variance). Identified
             novel variants (ie, rs1912461, rs61826952) were associated
             with differential central evoked theta power (loss - gain; P
             = .0037) and reward-related ventral striatum reactivity (P =
             .008), respectively. This study suggests that studying
             individual criteria may unveil new insights into the genetic
             etiology of AD liability.},
   Doi = {10.1111/gbb.12579},
   Key = {fds343721}
}

@article{fds348913,
   Author = {Elliott, ML and Belsky, DW and Anderson, K and Corcoran, DL and Ge, T and Knodt, A and Prinz, JA and Sugden, K and Williams, B and Ireland, D and Poulton, R and Caspi, A and Holmes, A and Moffitt, T and Hariri,
             AR},
   Title = {A Polygenic Score for Higher Educational Attainment is
             Associated with Larger Brains.},
   Journal = {Cerebral cortex (New York, N.Y. : 1991)},
   Volume = {29},
   Number = {8},
   Pages = {3496-3504},
   Year = {2019},
   Month = {July},
   Abstract = {People who score higher on intelligence tests tend to have
             larger brains. Twin studies suggest the same genetic factors
             influence both brain size and intelligence. This has led to
             the hypothesis that genetics influence intelligence partly
             by contributing to the development of larger brains. We
             tested this hypothesis using four large imaging genetics
             studies (combined N = 7965) with polygenic scores derived
             from a genome-wide association study (GWAS) of educational
             attainment, a correlate of intelligence. We conducted
             meta-analysis to test associations among participants'
             genetics, total brain volume (i.e., brain size), and
             cognitive test performance. Consistent with previous
             findings, participants with higher polygenic scores achieved
             higher scores on cognitive tests, as did participants with
             larger brains. Participants with higher polygenic scores
             also had larger brains. We found some evidence that brain
             size partly mediated associations between participants'
             education polygenic scores and their cognitive test
             performance. Effect sizes were larger in the
             population-based samples than in the convenience-based
             samples. Recruitment and retention of population-representative
             samples should be a priority for neuroscience research.
             Findings suggest promise for studies integrating GWAS
             discoveries with brain imaging to understand neurobiology
             linking genetics with cognitive performance.},
   Doi = {10.1093/cercor/bhy219},
   Key = {fds348913}
}

@article{fds364150,
   Author = {Avinun, R and Nevo, A and Hariri, AR},
   Title = {Genetic Risk for Rheumatoid Arthritis is Associated with
             Increased Striatal Volume in Healthy Young
             Adults.},
   Journal = {Scientific reports},
   Volume = {9},
   Number = {1},
   Pages = {10994},
   Year = {2019},
   Month = {July},
   Abstract = {Rheumatoid arthritis (RA), an autoimmune disease, has
             recently been associated with increased striatal volume and
             decreased intracranial volume (ICV) in longstanding
             patients. As inflammation has been shown to precede the
             clinical diagnosis of RA and it is a known moderator of
             neuro- and gliogenesis, we were interested in testing
             whether these brain morphological changes appear before the
             clinical onset of disease in healthy young adult volunteers,
             as a function of relative genetic risk for RA. Genetic and
             structural MRI data were available for 516 healthy
             non-Hispanic Caucasian university students (275 women, mean
             age 19.78 ± 1.24 years). Polygenic risk scores were
             computed for each individual based on a genome-wide
             association study of RA, so that higher scores indicated
             higher risk. Striatal volume (sum of caudate, putamen, and
             nucleus accumbens volumes) and ICV were derived for each
             individual from high-resolution T1-weighted images. After
             controlling for sex, age, genetic components of ethnicity,
             socioeconomic status, and depressive symptoms, we found that
             higher RA polygenic risk scores were associated with
             increased striatal volume, but not decreased ICV. Our
             findings suggest that increased striatal volume may be
             linked to processes that precede disease onset, such as
             inflammation, while decreased ICV may relate to disease
             progression.},
   Doi = {10.1038/s41598-019-47505-w},
   Key = {fds364150}
}

@article{fds364135,
   Author = {Wetherill, L and Lai, D and Johnson, EC and Anokhin, A and Bauer, L and Bucholz, KK and Dick, DM and Hariri, AR and Hesselbrock, V and Kamarajan, C and Kramer, J and Kuperman, S and Meyers, JL and Nurnberger, JI and Schuckit, M and Scott, DM and Taylor, RE and Tischfield, J and Porjesz, B and Goate, AM and Edenberg, HJ and Foroud,
             T and Bogdan, R and Agrawal, A},
   Title = {Genome-wide association study identifies loci associated
             with liability to alcohol and drug dependence that is
             associated with variability in reward-related ventral
             striatum activity in African- and European-Americans.},
   Journal = {Genes, brain, and behavior},
   Volume = {18},
   Number = {6},
   Pages = {e12580},
   Year = {2019},
   Month = {July},
   Abstract = {Genetic influences on alcohol and drug dependence partially
             overlap, however, specific loci underlying this overlap
             remain unclear. We conducted a genome-wide association study
             (GWAS) of a phenotype representing alcohol or illicit drug
             dependence (ANYDEP) among 7291 European-Americans (EA; 2927
             cases) and 3132 African-Americans (AA: 1315 cases)
             participating in the family-based Collaborative Study on the
             Genetics of Alcoholism. ANYDEP was heritable (h <sup>2</sup>
             in EA = 0.60, AA = 0.37). The AA GWAS identified three
             regions with genome-wide significant (GWS; P < 5E-08)
             single nucleotide polymorphisms (SNPs) on chromosomes 3
             (rs34066662, rs58801820) and 13 (rs75168521, rs78886294),
             and an insertion-deletion on chromosome 5 (chr5:141988181).
             No polymorphisms reached GWS in the EA. One GWS region
             (chromosome 1: rs1890881) emerged from a trans-ancestral
             meta-analysis (EA + AA) of ANYDEP, and was attributable
             to alcohol dependence in both samples. Four genes (AA: CRKL,
             DZIP3, SBK3; EA: P2RX6) and four sets of genes were
             significantly enriched within biological pathways for
             hemostasis and signal transduction. GWS signals did not
             replicate in two independent samples but there was weak
             evidence for association between rs1890881 and alcohol
             intake in the UK Biobank. Among 118 AA and 481 EA
             individuals from the Duke Neurogenetics Study, rs75168521
             and rs1890881 genotypes were associated with variability in
             reward-related ventral striatum activation. This study
             identified novel loci for substance dependence and provides
             preliminary evidence that these variants are also associated
             with individual differences in neural reward reactivity.
             Gene discovery efforts in non-European samples with distinct
             patterns of substance use may lead to the identification of
             novel ancestry-specific genetic markers of
             risk.},
   Doi = {10.1111/gbb.12580},
   Key = {fds364135}
}

@article{fds364151,
   Author = {Gerin, MI and Viding, E and Pingault, J-B and Puetz, VB and Knodt, AR and Radtke, SR and Brigidi, BD and Swartz, JR and Hariri, AR and McCrory,
             EJ},
   Title = {Heightened amygdala reactivity and increased stress
             generation predict internalizing symptoms in adults
             following childhood maltreatment.},
   Journal = {Journal of child psychology and psychiatry, and allied
             disciplines},
   Volume = {60},
   Number = {7},
   Pages = {752-761},
   Year = {2019},
   Month = {July},
   Abstract = {<h4>Background</h4>Childhood maltreatment is one of the most
             potent predictors of future psychopathology, including
             internalizing disorders. It remains unclear whether
             heightened amygdala reactivity to threat and elevated stress
             exposure may be implicated in the pathogenesis and
             maintenance of internalizing disorders among individuals
             with a history of childhood maltreatment.<h4>Methods</h4>Using
             data from a sample of 1,144 young adults, we investigated
             the contribution of baseline threat-related amygdala
             reactivity and prospective major stressful life events to
             internalizing symptoms severity 1 year later (on average)
             in individuals with a history of maltreatment (n = 100)
             and propensity score matched nonmaltreated peers
             (n = 96).<h4>Results</h4>Even after stringently matching
             for several potentially confounding variables - including
             baseline internalizing symptoms, socioeconomic status and IQ
             - childhood maltreatment status predicted increased amygdala
             reactivity at baseline, elevated post-baseline exposure to
             major stressful life events and internalizing symptoms at
             follow-up. We also showed, for the first time, that amygdala
             reactivity at baseline and also post-baseline exposure to
             major stressful life events mediated the association between
             a history of maltreatment and future internalizing
             symptoms.<h4>Conclusions</h4>These findings provide support
             for the view that maltreatment is a potent developmental
             insult leading to long-lasting neurocognitive recalibrations
             of the threat processing system. It is possible that such
             alterations, over time, may impact mental health functioning
             by compromising the ability to effectively negotiate
             everyday challenges (stress susceptibility). These
             alterations were not, however, found to sensitize an
             individual to the impact of major stressful life events. The
             results of this study also lend compelling support to the
             view that increased psychiatric risk, in the context of
             childhood maltreatment, follows from an increased propensity
             to experience major stressful life events (stress
             generation).},
   Doi = {10.1111/jcpp.13041},
   Key = {fds364151}
}

@article{fds364152,
   Author = {Farber, MJ and Romer, AL and Kim, MJ and Knodt, AR and Elsayed, NM and Williamson, DE and Hariri, AR},
   Title = {Paradoxical associations between familial affective
             responsiveness, stress, and amygdala reactivity.},
   Journal = {Emotion (Washington, D.C.)},
   Volume = {19},
   Number = {4},
   Pages = {645-654},
   Year = {2019},
   Month = {June},
   Abstract = {Studies of early life extremes such as trauma, abuse, and
             neglect highlight the critical importance of quality
             caregiving in the development of brain circuits supporting
             emotional behavior and mental health. The impact of
             normative variability in caregiving on such biobehavioral
             processes, however, is poorly understood. Here, we provide
             initial evidence that even subtle variability in normative
             caregiving maps onto individual differences in
             threat-related brain function and, potentially, associated
             psychopathology in adolescence. Specifically, we report that
             greater familial affective responsiveness is associated with
             heightened amygdala reactivity to interpersonal threat,
             particularly in adolescents having experienced relatively
             low recent stress. These findings extend the literature on
             the effects of caregiving extremes on brain function to
             subtle, normative variability but suggest that presumably
             protective factors may be associated with increased
             risk-related amygdala reactivity. We consider these
             paradoxical associations with regard to studies of basic
             associative threat learning and further consider their
             relevance for understanding potential effects of caregiving
             on psychological development. (PsycINFO Database Record (c)
             2019 APA, all rights reserved).},
   Doi = {10.1037/emo0000467},
   Key = {fds364152}
}

@article{fds364153,
   Author = {Kim, MJ and Elliott, ML and d'Arbeloff, TC and Knodt, AR and Radtke, SR and Brigidi, BD and Hariri, AR},
   Title = {Microstructural integrity of white matter moderates an
             association between childhood adversity and adult trait
             anger.},
   Journal = {Aggressive behavior},
   Volume = {45},
   Number = {3},
   Pages = {310-318},
   Year = {2019},
   Month = {May},
   Abstract = {Amongst a number of negative life sequelae associated with
             childhood adversity is the later expression of a higher
             dispositional tendency to experience anger and frustration
             to a wide range of situations (i.e., trait anger). We
             recently reported that an association between childhood
             adversity and trait anger is moderated by individual
             differences in both threat-related amygdala activity and
             executive control-related dorsolateral prefrontal cortex
             (dlPFC) activity, wherein individuals with relatively low
             amygdala and high dlPFC activity do not express higher trait
             anger even when having experienced childhood adversity.
             Here, we examine possible structural correlates of this
             functional dynamic using diffusion magnetic resonance
             imaging data from 647 young adult men and women volunteers.
             Specifically, we tested whether the degree of white matter
             microstructural integrity as indexed by fractional
             anisotropy modulated the association between childhood
             adversity and trait anger. Our analyses revealed that higher
             microstructural integrity of multiple pathways was
             associated with an attenuated link between childhood
             adversity and adult trait anger. Amongst these pathways was
             the uncinate fasciculus (UF; ΔR <sup>2</sup>  = 0.01),
             which not only provides a major anatomical link between the
             amygdala and prefrontal cortex but also is associated with
             individual differences in regulating negative emotion
             through top-down cognitive reappraisal. These findings
             suggest that higher microstructural integrity of distributed
             white matter pathways including but not limited to the UF
             may represent an anatomical foundation serving to buffer
             against the expression of childhood adversity as later trait
             anger, which is itself associated with multiple negative
             health outcomes.},
   Doi = {10.1002/ab.21820},
   Key = {fds364153}
}

@article{fds342490,
   Author = {Hariri, AR},
   Title = {The Emerging Importance of the Cerebellum in Broad Risk for
             Psychopathology.},
   Journal = {Neuron},
   Volume = {102},
   Number = {1},
   Pages = {17-20},
   Year = {2019},
   Month = {April},
   Abstract = {Recent research has identified a single factor accounting
             for broad risk to experience common forms of
             psychopathology. Structural alterations of cerebellar
             circuitry have emerged as a neural nexus of this broad risk,
             highlighting the cerebellum's importance for executive
             control.},
   Doi = {10.1016/j.neuron.2019.02.031},
   Key = {fds342490}
}

@article{fds364182,
   Author = {Elliott, ML and Knodt, AR and Cooke, M and Kim, MJ and Melzer, TR and Keenan, R and Ireland, D and Ramrakha, S and Poulton, R and Caspi, A and Moffitt, TE and Hariri, AR},
   Title = {General functional connectivity: Shared features of
             resting-state and task fMRI drive reliable and heritable
             individual differences in functional brain
             networks.},
   Journal = {NeuroImage},
   Volume = {189},
   Pages = {516-532},
   Year = {2019},
   Month = {April},
   Abstract = {Intrinsic connectivity, measured using resting-state fMRI,
             has emerged as a fundamental tool in the study of the human
             brain. However, due to practical limitations, many studies
             do not collect enough resting-state data to generate
             reliable measures of intrinsic connectivity necessary for
             studying individual differences. Here we present general
             functional connectivity (GFC) as a method for leveraging
             shared features across resting-state and task fMRI and
             demonstrate in the Human Connectome Project and the Dunedin
             Study that GFC offers better test-retest reliability than
             intrinsic connectivity estimated from the same amount of
             resting-state data alone. Furthermore, at equivalent scan
             lengths, GFC displayed higher estimates of heritability than
             resting-state functional connectivity. We also found that
             predictions of cognitive ability from GFC generalized across
             datasets, performing as well or better than resting-state or
             task data alone. Collectively, our work suggests that GFC
             can improve the reliability of intrinsic connectivity
             estimates in existing datasets and, subsequently, the
             opportunity to identify meaningful correlates of individual
             differences in behavior. Given that task and resting-state
             data are often collected together, many researchers can
             immediately derive more reliable measures of intrinsic
             connectivity through the adoption of GFC rather than solely
             using resting-state data. Moreover, by better capturing
             heritable variation in intrinsic connectivity, GFC
             represents a novel endophenotype with broad applications in
             clinical neuroscience and biomarker discovery.},
   Doi = {10.1016/j.neuroimage.2019.01.068},
   Key = {fds364182}
}

@article{fds364154,
   Author = {Romer, AL and Su Kang and M and Nikolova, YS and Gearhardt, AN and Hariri,
             AR},
   Title = {Dopamine genetic risk is related to food addiction and body
             mass through reduced reward-related ventral striatum
             activity.},
   Journal = {Appetite},
   Volume = {133},
   Pages = {24-31},
   Year = {2019},
   Month = {February},
   Abstract = {The prevalence rate of obesity continues to rise in the
             U.S., but effective treatment options remain elusive
             resulting in increased emphasis on prevention. One such area
             of prevention research capitalizes on the relatively novel
             behavioral construct of food addiction, which has been
             implicated in obesity. Food addiction reflects an
             individual's propensity for compulsive eating despite
             negative consequences, and shares not only symptoms with
             both eating and substance use disorders but also genetic and
             neural correlates within neural reward-circuitry modulated
             by dopamine. Here, we examined associations between food
             addiction scores, body mass index (BMI), reward-related
             ventral striatum activity, and a polygenic score
             approximating dopamine signaling in 115 non-Hispanic
             Caucasian young adult university students. As predicted,
             polygenic dopamine scores were related to ventral striatum
             activity, which in turn was associated with higher food
             addiction scores. In addition, food addiction was related to
             BMI. An exploratory post-hoc path analysis further indicated
             that polygenic scores were indirectly related to both food
             addiction and BMI, in part, through ventral striatum
             activity. Collectively, our results provide evidence
             supporting the utility of food addiction in weight gain
             prevention research by establishing links with known
             risk-related neural and genetic biomarkers.},
   Doi = {10.1016/j.appet.2018.09.010},
   Key = {fds364154}
}

@article{fds346873,
   Author = {Kim, MJ and Farber, MJ and Knodt, AR and Hariri, AR},
   Title = {Corticolimbic circuit structure moderates an association
             between early life stress and later trait
             anxiety.},
   Journal = {NeuroImage. Clinical},
   Volume = {24},
   Pages = {102050},
   Year = {2019},
   Month = {January},
   Abstract = {Childhood adversity is associated with a wide range of
             negative behavioral and neurodevelopmental consequences.
             However, individuals vary substantially in their sensitivity
             to such adversity. Here, we examined how individual
             variability in structural features of the corticolimbic
             circuit, which plays a key role in emotional reactivity,
             moderates the association between childhood adversity and
             later trait anxiety in 798 young adult university students.
             Consistent with prior research, higher self-reported
             childhood adversity was significantly associated with higher
             self-reported trait anxiety. However, this association was
             attenuated in participants with higher microstructural
             integrity of the uncinate fasciculus and greater thickness
             of the orbitofrontal cortex. These structural properties of
             the corticolimbic circuit may capture a neural profile of
             relative resiliency to early life stress, especially against
             the negative effects of childhood adversity on later trait
             anxiety.},
   Doi = {10.1016/j.nicl.2019.102050},
   Key = {fds346873}
}

@article{fds366148,
   Author = {d'Arbeloff, T and Elliott, ML and Knodt, AR and Melzer, TR and Keenan,
             R and Ireland, D and Ramrakha, S and Poulton, R and Anderson, T and Caspi,
             A and Moffitt, TE and Hariri, AR},
   Title = {White matter hyperintensities are common in midlife and
             already associated with cognitive decline.},
   Journal = {Brain communications},
   Volume = {1},
   Number = {1},
   Pages = {fcz041},
   Year = {2019},
   Month = {January},
   Abstract = {White matter hyperintensities proliferate as the brain ages
             and are associated with increased risk for cognitive decline
             as well as Alzheimer's disease and related dementias. As
             such, white matter hyperintensities have been targeted as a
             surrogate biomarker in intervention trials with older
             adults. However, it is unclear at what stage of aging white
             matter hyperintensities begin to relate to cognition and if
             they may be a viable target for early prevention. In the
             Dunedin Study, a population-representative cohort followed
             since birth, we measured white matter hyperintensities in
             843 45-year-old participants using T<sub>2</sub>-weighted
             magnetic resonance imaging and we assessed cognitive decline
             from childhood to midlife. We found that white matter
             hyperintensities were common at age 45 and that white matter
             hyperintensity volume was modestly associated with both
             lower childhood (<i>ß</i> = -0.08, <i>P </i>=<i> </i>0.013)
             and adult IQ (<i>ß</i>=-0.15, <i>P </i><<i> </i>0.001).
             Moreover, white matter hyperintensity volume was associated
             with greater cognitive decline from childhood to midlife
             (<i>ß</i>=-0.09, <i>P </i><<i> </i>0.001). Our results
             demonstrate that a link between white matter
             hyperintensities and early signs of cognitive decline is
             detectable decades before clinical symptoms of dementia
             emerge. Thus, white matter hyperintensities may be a useful
             surrogate biomarker for identifying individuals in midlife
             at risk for future accelerated cognitive decline and
             selecting participants for dementia prevention
             trials.},
   Doi = {10.1093/braincomms/fcz041},
   Key = {fds366148}
}

@article{fds364132,
   Author = {Knodt, AR and Burke, JR and Welsh-Bohmer, KA and Plassman, BL and Burns,
             DK and Brannan, SK and Kukulka, M and Wu, J and Hariri,
             AR},
   Title = {Effects of pioglitazone on mnemonic hippocampal function: A
             blood oxygen level-dependent functional magnetic resonance
             imaging study in elderly adults.},
   Journal = {Alzheimers Dement (N Y)},
   Volume = {5},
   Pages = {254-263},
   Year = {2019},
   Abstract = {INTRODUCTION: Mitochondrial dysfunction is implicated in the
             pathophysiology of Alzheimer's disease (AD). Accordingly,
             drugs that positively influence mitochondrial function are
             being evaluated in delay-of-onset clinical trials with
             at-risk individuals. Such ongoing clinical research can be
             advanced by developing a better understanding of how these
             drugs affect intermediate brain phenotypes associated with
             both AD risk and pathophysiology. METHODS: Using a
             randomized, parallel-group, placebo-controlled design in 55
             healthy elderly volunteers, we explored the effects of oral,
             low-dose pioglitazone, a thiazolidinedione with
             promitochondrial effects, on hippocampal activity measured
             with functional magnetic resonance imaging during the
             encoding of novel face-name pairs. RESULTS: Compared with
             placebo, 0.6 mg of pioglitazone (but not 2.1 mg, 3.9 mg,
             or 6.0 mg) administered daily for 14 days was associated
             with significant increases in right hippocampal activation
             during encoding of novel face-name pairs at day 7 and day
             14, relative to baseline. DISCUSSION: Our exploratory
             analyses suggest that low-dose pioglitazone has measurable
             effects on mnemonic brain function associated with AD risk
             and pathophysiology.},
   Doi = {10.1016/j.trci.2019.05.004},
   Key = {fds364132}
}

@article{fds335689,
   Author = {Sethi, A and McCrory, E and Puetz, V and Hoffmann, F and Knodt, AR and Radtke, SR and Brigidi, BD and Hariri, AR and Viding,
             E},
   Title = {Primary and Secondary Variants of Psychopathy in a Volunteer
             Sample Are Associated With Different Neurocognitive
             Mechanisms.},
   Journal = {Biological psychiatry. Cognitive neuroscience and
             neuroimaging},
   Volume = {3},
   Number = {12},
   Pages = {1013-1021},
   Publisher = {Elsevier BV},
   Year = {2018},
   Month = {December},
   Abstract = {<h4>Background</h4>Recent work has indicated that there at
             least two distinct subtypes of psychopathy. Primary
             psychopathy is characterized by low anxiety and thought to
             result from a genetic predisposition, whereas secondary
             psychopathy is characterized by high anxiety and thought to
             develop in response to environmental adversity. Primary
             psychopathy is robustly associated with reduced neural
             activation to others' emotions and, in particular, distress.
             However, it has been proposed that the secondary
             presentation has different neurocognitive
             correlates.<h4>Methods</h4>Primary (n = 50), secondary
             (n = 100), and comparison (n = 82) groups were drawn from
             a large volunteer sample (N = 1444) using a quartile-split
             approach across psychopathic trait (affective-interpersonal)
             and anxiety measures. Participants performed a widely
             utilized emotional face processing task during functional
             magnetic resonance imaging.<h4>Results</h4>The primary group
             showed reduced amygdala and insula activity in response to
             fear. The secondary group did not differ from the comparison
             group in these regions. Instead, the secondary group showed
             reduced activity compared with the comparison group in other
             areas, including the superior temporal sulcus/inferior
             parietal lobe, thalamus, pallidum, and substantia nigra.
             Both psychopathy groups also showed reduced activity in
             response to fear in the anterior cingulate cortex. During
             anger processing, the secondary group exhibited reduced
             activity in the anterior cingulate cortex compared with the
             primary group.<h4>Conclusions</h4>Distinct neural correlates
             of fear processing characterize individuals with primary and
             secondary psychopathy. The reduced neural response to fear
             that characterizes individuals with the primary variant of
             psychopathic traits is not observed in individuals with the
             secondary presentation. The neurocognitive mechanisms
             underpinning secondary psychopathy warrant further
             systematic investigation.},
   Doi = {10.1016/j.bpsc.2018.04.002},
   Key = {fds335689}
}

@article{fds335682,
   Author = {Lam, M and Trampush, JW and Yu, J and Knowles, E and Djurovic, S and Melle,
             I and Sundet, K and Christoforou, A and Reinvang, I and DeRosse, P and Lundervold, AJ and Steen, VM and Espeseth, T and Räikkönen, K and Widen, E and Palotie, A and Eriksson, JG and Giegling, I and Konte, B and Roussos, P and Giakoumaki, S and Burdick, KE and Payton, A and Ollier,
             W and Chiba-Falek, O and Attix, DK and Need, AC and Cirulli, ET and Voineskos, AN and Stefanis, NC and Avramopoulos, D and Hatzimanolis,
             A and Arking, DE and Smyrnis, N and Bilder, RM and Freimer, NA and Cannon,
             TD and London, E and Poldrack, RA and Sabb, FW and Congdon, E and Conley,
             ED and Scult, MA and Dickinson, D and Straub, RE and Donohoe, G and Morris,
             D and Corvin, A and Gill, M and Hariri, AR and Weinberger, DR and Pendleton, N and Bitsios, P and Rujescu, D and Lahti, J and Hellard, SL and Keller, MC and Andreassen, OA and Glahn, DC and Malhotra, AK and Lencz,
             T},
   Title = {Multi-Trait Analysis of GWAS and Biological Insights Into
             Cognition: A Response to Hill (2018).},
   Journal = {Twin Res Hum Genet},
   Volume = {21},
   Number = {5},
   Pages = {394-397},
   Year = {2018},
   Month = {October},
   Abstract = {Hill (Twin Research and Human Genetics, Vol. 21, 2018,
             84-88) presented a critique of our recently published paper
             in Cell Reports entitled 'Large-Scale Cognitive GWAS
             Meta-Analysis Reveals Tissue-Specific Neural Expression and
             Potential Nootropic Drug Targets' (Lam et al., Cell Reports,
             Vol. 21, 2017, 2597-2613). Specifically, Hill offered
             several interrelated comments suggesting potential problems
             with our use of a new analytic method called Multi-Trait
             Analysis of GWAS (MTAG) (Turley et al., Nature Genetics,
             Vol. 50, 2018, 229-237). In this brief article, we respond
             to each of these concerns. Using empirical data, we conclude
             that our MTAG results do not suffer from 'inflation in the
             FDR [false discovery rate]', as suggested by Hill (Twin
             Research and Human Genetics, Vol. 21, 2018, 84-88), and are
             not 'more relevant to the genetic contributions to education
             than they are to the genetic contributions to
             intelligence'.},
   Doi = {10.1017/thg.2018.46},
   Key = {fds335682}
}

@article{fds335686,
   Author = {d'Arbeloff, TC and Kim, MJ and Knodt, AR and Radtke, SR and Brigidi, BD and Hariri, AR},
   Title = {Microstructural integrity of a pathway connecting the
             prefrontal cortex and amygdala moderates the association
             between cognitive reappraisal and negative
             emotions.},
   Journal = {Emotion (Washington, D.C.)},
   Volume = {18},
   Number = {6},
   Pages = {912-915},
   Year = {2018},
   Month = {September},
   Abstract = {Cognitive reappraisal is a commonly used form of emotion
             regulation that utilizes frontal-executive control to
             reframe an approaching emotional event to moderate its
             potential psychological impact. Use of cognitive reappraisal
             has been associated with diminished experience of anxiety
             and depressive symptoms, as well as greater overall
             well-being. Using data from a study of 647 healthy young
             adults, we provide initial evidence that an association
             between typical use of cognitive reappraisal in daily life
             and the experience of anxiety and depressive symptoms is
             moderated by the microstructural integrity of the uncinate
             fasciculus, which provides a major anatomical link between
             the amygdala and prefrontal cortex. Our findings are
             consistent with the nature of top-down regulation of
             bottom-up negative emotions and suggest the uncinate
             fasciculus may be a useful target in the search for
             biomarkers predicting not only disorder risk but also
             response to psychotherapy utilizing cognitive reappraisal.
             (PsycINFO Database Record},
   Doi = {10.1037/emo0000447},
   Key = {fds335686}
}

@article{fds332980,
   Author = {Hanson, JL and Knodt, AR and Brigidi, BD and Hariri,
             AR},
   Title = {Heightened connectivity between the ventral striatum and
             medial prefrontal cortex as a biomarker for stress-related
             psychopathology: understanding interactive effects of early
             and more recent stress.},
   Journal = {Psychological medicine},
   Volume = {48},
   Number = {11},
   Pages = {1835-1843},
   Year = {2018},
   Month = {August},
   Abstract = {<h4>Background</h4>The experience of childhood maltreatment
             is a significant risk factor for the development of
             depression. This risk is particularly heightened after
             exposure to additional, more contemporaneous stress. While
             behavioral evidence exists for this relation, little is
             known about biological correlates of these stress
             interactions. Identifying such correlates may provide
             biomarkers of risk for later depression.<h4>Methods</h4>Here,
             we leverage behavioral, experiential, and neuroimaging data
             from the Duke Neurogenetics Study to identify potential
             biomarkers of stress exposure. Based on the past research,
             we were specifically interested in reward-related
             connectivity and the interaction of early and more recent
             stress. We examined psychophysiological interactions between
             the ventral striatum and other brain regions in relation to
             these stress variables, as well as measures of internalizing
             symptomatology (n = 926, participant age range = 18-22 years
             of age).<h4>Results</h4>We found relatively increased
             reward-related functional connectivity between the left
             ventral striatum and the medial prefrontal cortex in
             individuals exposed to greater levels of childhood
             maltreatment who also experienced greater levels of recent
             life stress (β = 0.199, p < 0.005). This pattern of
             functional connectivity was further associated with elevated
             symptoms of depression (β = 0.089, p = 0.006). Furthermore,
             using a moderated mediation framework, we demonstrate that
             this functional connectivity provides a biological link
             between cumulative stress exposure and internalizing
             symptomatology.<h4>Conclusions</h4>These findings suggest a
             novel biomarker linking cumulative stress exposure with the
             later experience of depressive symptoms. Our results are
             discussed in the context of past research examining stress
             exposure in relation to depression.},
   Doi = {10.1017/s0033291717003348},
   Key = {fds332980}
}

@article{fds364155,
   Author = {Elsayed, NM and Kim, MJ and Fields, KM and Olvera, RL and Hariri, AR and Williamson, DE},
   Title = {Trajectories of Alcohol Initiation and Use During
             Adolescence: The Role of Stress and Amygdala
             Reactivity.},
   Journal = {Journal of the American Academy of Child and Adolescent
             Psychiatry},
   Volume = {57},
   Number = {8},
   Pages = {550-560},
   Year = {2018},
   Month = {August},
   Abstract = {<h4>Objective</h4>Early alcohol use initiation predicts
             onset of alcohol use disorders in adulthood. However, little
             is known about developmental trajectories of alcohol use
             initiation and their putative biological and environmental
             correlates.<h4>Method</h4>Adolescents (N = 330) with high
             or low familial loading for depression were assessed
             annually for up to 6 years. Data were collected assessing
             affective symptoms, alcohol use, and stress at each
             assessment. Adolescents also participated in a functional
             magnetic resonance imaging protocol that included
             measurement of threat-related amygdala and reward-related
             ventral striatum activity.<h4>Results</h4>Latent class
             analyses identified 2 trajectories of alcohol use
             initiation. Early initiators (n = 32) reported greater
             baseline alcohol use and rate of change of use compared with
             late initiators and/or current abstainers (n = 298). Early
             initiators reported higher baseline levels of stressful life
             events (p = .001) and exhibited higher amygdala (p = .001)
             but not ventral striatum activity compared with late
             initiators. Early initiators were 15.3 times more likely to
             have a full drink (p < .0001), 9.1 times more likely to
             experience intoxication (p < .0001), and 6.7 times more
             likely to develop an alcohol use disorder by 19 years of age
             compared with late initiators (p = .003).<h4>Conclusion</h4>Adolescents
             on a trajectory of early alcohol use initiation have higher
             levels of stress, have increased threat-related amygdala
             activity, are more likely to consume a full standard
             alcoholic drink, are more likely to experience early
             intoxication, and are at a heightened risk for the onset of
             an alcohol use disorder.},
   Doi = {10.1016/j.jaac.2018.05.011},
   Key = {fds364155}
}

@article{fds335683,
   Author = {Kim, MJ and Scult, MA and Knodt, AR and Radtke, SR and d'Arbeloff, TC and Brigidi, BD and Hariri, AR},
   Title = {A Link Between Childhood Adversity and Trait Anger Reflects
             Relative Activity of the Amygdala and Dorsolateral
             Prefrontal Cortex.},
   Journal = {Biological psychiatry. Cognitive neuroscience and
             neuroimaging},
   Volume = {3},
   Number = {7},
   Pages = {644-649},
   Year = {2018},
   Month = {July},
   Abstract = {<h4>Background</h4>Trait anger, or the dispositional
             tendency to experience a wide range of situations as
             annoying or frustrating, is associated with negative mental
             and physical health outcomes. The experience of adversity
             during childhood is one risk factor for the later emergence
             of high trait anger. This association has been hypothesized
             to reflect alterations in neural circuits supporting
             bottom-up threat processing and top-down executive
             control.<h4>Methods</h4>Here, using functional magnetic
             resonance imaging and self-report questionnaire data from
             220 volunteers, we examined how individual differences in
             top-down prefrontal executive control and bottom-up amygdala
             threat activity modulate the association between childhood
             adversity and trait anger during young adulthood.<h4>Results</h4>We
             report that the association between childhood adversity and
             trait anger is attenuated specifically in young adults who
             have both relatively low threat-related amygdala activity
             and high executive control-related dorsolateral prefrontal
             cortex activity.<h4>Conclusions</h4>These brain activity
             patterns suggest that simultaneous consideration of their
             underlying cognitive processes-namely, threat processing and
             executive control-may be useful in strategies designed to
             mitigate the negative mental health consequences of
             childhood adversity.},
   Doi = {10.1016/j.bpsc.2018.03.006},
   Key = {fds335683}
}

@article{fds335684,
   Author = {Savage, JE and Jansen, PR and Stringer, S and Watanabe, K and Bryois, J and de Leeuw, CA and Nagel, M and Awasthi, S and Barr, PB and Coleman, JRI and Grasby, KL and Hammerschlag, AR and Kaminski, JA and Karlsson, R and Krapohl, E and Lam, M and Nygaard, M and Reynolds, CA and Trampush, JW and Young, H and Zabaneh, D and Hägg, S and Hansell, NK and Karlsson, IK and Linnarsson, S and Montgomery, GW and Muñoz-Manchado, AB and Quinlan,
             EB and Schumann, G and Skene, NG and Webb, BT and White, T and Arking, DE and Avramopoulos, D and Bilder, RM and Bitsios, P and Burdick, KE and Cannon, TD and Chiba-Falek, O and Christoforou, A and Cirulli, ET and Congdon, E and Corvin, A and Davies, G and Deary, IJ and DeRosse, P and Dickinson, D and Djurovic, S and Donohoe, G and Conley, ED and Eriksson,
             JG and Espeseth, T and Freimer, NA and Giakoumaki, S and Giegling, I and Gill, M and Glahn, DC and Hariri, AR and Hatzimanolis, A and Keller, MC and Knowles, E and Koltai, D and Konte, B and Lahti, J and Le Hellard and S and Lencz, T and Liewald, DC and London, E and Lundervold, AJ and Malhotra,
             AK and Melle, I and Morris, D and Need, AC and Ollier, W and Palotie, A and Payton, A and Pendleton, N and Poldrack, RA and Räikkönen, K and Reinvang, I and Roussos, P and Rujescu, D and Sabb, FW and Scult, MA and Smeland, OB and Smyrnis, N and Starr, JM and Steen, VM and Stefanis, NC and Straub, RE and Sundet, K and Tiemeier, H and Voineskos, AN and Weinberger, DR and Widen, E and Yu, J and Abecasis, G and Andreassen,
             OA and Breen, G and Christiansen, L and Debrabant, B and Dick, DM and Heinz, A and Hjerling-Leffler, J and Ikram, MA and Kendler, KS and Martin, NG and Medland, SE and Pedersen, NL and Plomin, R and Polderman,
             TJC and Ripke, S and van der Sluis, S and Sullivan, PF and Vrieze, SI and Wright, MJ and Posthuma, D},
   Title = {Genome-wide association meta-analysis in 269,867 individuals
             identifies new genetic and functional links to
             intelligence.},
   Journal = {Nat Genet},
   Volume = {50},
   Number = {7},
   Pages = {912-919},
   Year = {2018},
   Month = {July},
   Abstract = {Intelligence is highly heritable1 and a major determinant of
             human health and well-being2. Recent genome-wide
             meta-analyses have identified 24 genomic loci linked to
             variation in intelligence3-7, but much about its genetic
             underpinnings remains to be discovered. Here, we present a
             large-scale genetic association study of intelligence
             (n = 269,867), identifying 205 associated genomic loci
             (190 new) and 1,016 genes (939 new) via positional mapping,
             expression quantitative trait locus (eQTL) mapping,
             chromatin interaction mapping, and gene-based association
             analysis. We find enrichment of genetic effects in conserved
             and coding regions and associations with 146 nonsynonymous
             exonic variants. Associated genes are strongly expressed in
             the brain, specifically in striatal medium spiny neurons and
             hippocampal pyramidal neurons. Gene set analyses implicate
             pathways related to nervous system development and synaptic
             structure. We confirm previous strong genetic correlations
             with multiple health-related outcomes, and Mendelian
             randomization analysis results suggest protective effects of
             intelligence for Alzheimer's disease and ADHD and
             bidirectional causation with pleiotropic effects for
             schizophrenia. These results are a major step forward in
             understanding the neurobiology of cognitive function as well
             as genetically related neurological and psychiatric
             disorders.},
   Doi = {10.1038/s41588-018-0152-6},
   Key = {fds335684}
}

@article{fds331566,
   Author = {Avinun, R and Nevo, A and Knodt, AR and Elliott, ML and Hariri,
             AR},
   Title = {Replication in Imaging Genetics: The Case of Threat-Related
             Amygdala Reactivity.},
   Journal = {Biological psychiatry},
   Volume = {84},
   Number = {2},
   Pages = {148-159},
   Year = {2018},
   Month = {July},
   Abstract = {<h4>Background</h4>Low replication rates are a concern in
             most, if not all, scientific disciplines. In psychiatric
             genetics specifically, targeting intermediate brain
             phenotypes, which are more closely associated with putative
             genetic effects, was touted as a strategy leading to
             increased power and replicability. In the current study, we
             attempted to replicate previously published associations
             between single nucleotide polymorphisms and threat-related
             amygdala reactivity, which represents a robust brain
             phenotype not only implicated in the pathophysiology of
             multiple disorders, but also used as a biomarker of future
             risk.<h4>Methods</h4>We conducted a literature search for
             published associations between single nucleotide
             polymorphisms and threat-related amygdala reactivity and
             found 37 unique findings. Our replication sample consisted
             of 1117 young adult volunteers (629 women, mean age 19.72 ±
             1.25 years) for whom both genetic and functional magnetic
             resonance imaging data were available.<h4>Results</h4>Of the
             37 unique associations identified, only three replicated as
             previously reported. When exploratory analyses were
             conducted with different model parameters compared to the
             original findings, significant associations were identified
             for 28 additional studies: eight of these were for a
             different contrast/laterality; five for a different gender
             and/or race/ethnicity; and 15 in the opposite direction and
             for a different contrast, laterality, gender, and/or
             race/ethnicity. No significant associations, regardless of
             model parameters, were detected for six studies. Notably,
             none of the significant associations survived correction for
             multiple comparisons.<h4>Conclusions</h4>We discuss these
             patterns of poor replication with regard to the general
             strategy of targeting intermediate brain phenotypes in
             genetic association studies and the growing importance of
             advancing the replicability of imaging genetics
             findings.},
   Doi = {10.1016/j.biopsych.2017.11.010},
   Key = {fds331566}
}

@article{fds335691,
   Author = {Miller, JA and Scult, MA and Conley, ED and Chen, Q and Weinberger, DR and Hariri, AR},
   Title = {Effects of Schizophrenia Polygenic Risk Scores on Brain
             Activity and Performance During Working Memory Subprocesses
             in Healthy Young Adults.},
   Journal = {Schizophrenia bulletin},
   Volume = {44},
   Number = {4},
   Pages = {844-853},
   Year = {2018},
   Month = {June},
   Abstract = {Recent work has begun to shed light on the neural correlates
             and possible mechanisms of polygenic risk for schizophrenia.
             Here, we map a schizophrenia polygenic risk profile score
             (PRS) based on genome-wide association study significant
             loci onto variability in the activity and functional
             connectivity of a frontoparietal network supporting the
             manipulation versus maintenance of information during a
             numerical working memory (WM) task in healthy young adults
             (n = 99, mean age = 19.8). Our analyses revealed that higher
             PRS was associated with hypoactivity of the dorsolateral
             prefrontal cortex (dlPFC) during the manipulation but not
             maintenance of information in WM (r2 = .0576, P = .018).
             Post hoc analyses revealed that PRS-modulated dlPFC
             hypoactivity correlated with faster reaction times during WM
             manipulation (r2 = .0967, P = .002), and faster processing
             speed (r2 = .0967, P = .003) on a separate behavioral task.
             These PRS-associated patterns recapitulate dlPFC
             hypoactivity observed in patients with schizophrenia during
             central executive manipulation of information in WM on this
             task.},
   Doi = {10.1093/schbul/sbx140},
   Key = {fds335691}
}

@article{fds335685,
   Author = {Davies, G and Lam, M and Harris, SE and Trampush, JW and Luciano, M and Hill, WD and Hagenaars, SP and Ritchie, SJ and Marioni, RE and Fawns-Ritchie, C and Liewald, DCM and Okely, JA and Ahola-Olli, AV and Barnes, CLK and Bertram, L and Bis, JC and Burdick, KE and Christoforou,
             A and DeRosse, P and Djurovic, S and Espeseth, T and Giakoumaki, S and Giddaluru, S and Gustavson, DE and Hayward, C and Hofer, E and Ikram,
             MA and Karlsson, R and Knowles, E and Lahti, J and Leber, M and Li, S and Mather, KA and Melle, I and Morris, D and Oldmeadow, C and Palviainen,
             T and Payton, A and Pazoki, R and Petrovic, K and Reynolds, CA and Sargurupremraj, M and Scholz, M and Smith, JA and Smith, AV and Terzikhan, N and Thalamuthu, A and Trompet, S and van der Lee, SJ and Ware, EB and Windham, BG and Wright, MJ and Yang, J and Yu, J and Ames, D and Amin, N and Amouyel, P and Andreassen, OA and Armstrong, NJ and Assareh,
             AA and Attia, JR and Attix, D and Avramopoulos, D and Bennett, DA and Böhmer, AC and Boyle, PA and Brodaty, H and Campbell, H and Cannon, TD and Cirulli, ET and Congdon, E and Conley, ED and Corley, J and Cox, SR and Dale, AM and Dehghan, A and Dick, D and Dickinson, D and Eriksson, JG and Evangelou, E and Faul, JD and Ford, I and Freimer, NA and Gao, H and Giegling, I and Gillespie, NA and Gordon, SD and Gottesman, RF and Griswold, ME and Gudnason, V and Harris, TB and Hartmann, AM and Hatzimanolis, A and Heiss, G and Holliday, EG and Joshi, PK and Kähönen, M and Kardia, SLR and Karlsson, I and Kleineidam, L and Knopman, DS and Kochan, NA and Konte, B and Kwok, JB and Le Hellard and S and Lee, T and Lehtimäki, T and Li, S-C and Lill, CM and Liu, T and Koini, M and London, E and Longstreth, WT and Lopez, OL and Loukola, A and Luck, T and Lundervold, AJ and Lundquist, A and Lyytikäinen, L-P and Martin, NG and Montgomery, GW and Murray, AD and Need, AC and Noordam, R and Nyberg, L and Ollier, W and Papenberg, G and Pattie, A and Polasek, O and Poldrack,
             RA and Psaty, BM and Reppermund, S and Riedel-Heller, SG and Rose, RJ and Rotter, JI and Roussos, P and Rovio, SP and Saba, Y and Sabb, FW and Sachdev, PS and Satizabal, CL and Schmid, M and Scott, RJ and Scult, MA and Simino, J and Slagboom, PE and Smyrnis, N and Soumaré, A and Stefanis,
             NC and Stott, DJ and Straub, RE and Sundet, K and Taylor, AM and Taylor,
             KD and Tzoulaki, I and Tzourio, C and Uitterlinden, A and Vitart, V and Voineskos, AN and Kaprio, J and Wagner, M and Wagner, H and Weinhold, L and Wen, KH and Widen, E and Yang, Q and Zhao, W and Adams, HHH and Arking, DE and Bilder, RM and Bitsios, P and Boerwinkle, E and Chiba-Falek, O and Corvin, A and De Jager and PL and Debette, S and Donohoe, G and Elliott, P and Fitzpatrick, AL and Gill, M and Glahn, DC and Hägg, S and Hansell, NK and Hariri, AR and Ikram, MK and Jukema, JW and Vuoksimaa, E and Keller, MC and Kremen, WS and Launer, L and Lindenberger, U and Palotie, A and Pedersen, NL and Pendleton, N and Porteous, DJ and Räikkönen, K and Raitakari, OT and Ramirez, A and Reinvang, I and Rudan, I and Dan
             Rujescu, and Schmidt, R and Schmidt, H and Schofield, PW and Schofield,
             PR and Starr, JM and Steen, VM and Trollor, JN and Turner, ST and Van
             Duijn, CM and Villringer, A and Weinberger, DR and Weir, DR and Wilson,
             JF and Malhotra, A and McIntosh, AM and Gale, CR and Seshadri, S and Mosley, TH and Bressler, J and Lencz, T and Deary,
             IJ},
   Title = {Study of 300,486 individuals identifies 148 independent
             genetic loci influencing general cognitive
             function.},
   Journal = {Nat Commun},
   Volume = {9},
   Number = {1},
   Pages = {2098},
   Year = {2018},
   Month = {May},
   Abstract = {General cognitive function is a prominent and relatively
             stable human trait that is associated with many important
             life outcomes. We combine cognitive and genetic data from
             the CHARGE and COGENT consortia, and UK Biobank (total
             N = 300,486; age 16-102) and find 148 genome-wide
             significant independent loci (P < 5 × 10-8)
             associated with general cognitive function. Within the novel
             genetic loci are variants associated with neurodegenerative
             and neurodevelopmental disorders, physical and psychiatric
             illnesses, and brain structure. Gene-based analyses find 709
             genes associated with general cognitive function. Expression
             levels across the cortex are associated with general
             cognitive function. Using polygenic scores, up to 4.3% of
             variance in general cognitive function is predicted in
             independent samples. We detect significant genetic overlap
             between general cognitive function, reaction time, and many
             health variables including eyesight, hypertension, and
             longevity. In conclusion we identify novel genetic loci and
             pathways contributing to the heritability of general
             cognitive function.},
   Doi = {10.1038/s41467-018-04362-x},
   Key = {fds335685}
}

@article{fds335687,
   Author = {Agrawal, A and Chou, Y-L and Carey, CE and Baranger, DAA and Zhang, B and Sherva, R and Wetherill, L and Kapoor, M and Wang, J-C and Bertelsen, S and Anokhin, AP and Hesselbrock, V and Kramer, J and Lynskey, MT and Meyers,
             JL and Nurnberger, JI and Rice, JP and Tischfield, J and Bierut, LJ and Degenhardt, L and Farrer, LA and Gelernter, J and Hariri, AR and Heath,
             AC and Kranzler, HR and Madden, PAF and Martin, NG and Montgomery, GW and Porjesz, B and Wang, T and Whitfield, JB and Edenberg, HJ and Foroud, T and Goate, AM and Bogdan, R and Nelson, EC},
   Title = {Genome-wide association study identifies a novel locus for
             cannabis dependence.},
   Journal = {Molecular psychiatry},
   Volume = {23},
   Number = {5},
   Pages = {1293-1302},
   Year = {2018},
   Month = {May},
   Abstract = {Despite moderate heritability, only one study has identified
             genome-wide significant loci for cannabis-related
             phenotypes. We conducted meta-analyses of genome-wide
             association study data on 2080 cannabis-dependent cases and
             6435 cannabis-exposed controls of European descent. A
             cluster of correlated single-nucleotide polymorphisms (SNPs)
             in a novel region on chromosome 10 was genome-wide
             significant (lowest P=1.3E-8). Among the SNPs, rs1409568
             showed enrichment for H3K4me1 and H3K427ac marks, suggesting
             its role as an enhancer in addiction-relevant brain regions,
             such as the dorsolateral prefrontal cortex and the angular
             and cingulate gyri. This SNP is also predicted to modify
             binding scores for several transcription factors. We found
             modest evidence for replication for rs1409568 in an
             independent cohort of African American (896 cases and 1591
             controls; P=0.03) but not European American (EA; 781 cases
             and 1905 controls) participants. The combined meta-analysis
             (3757 cases and 9931 controls) indicated trend-level
             significance for rs1409568 (P=2.85E-7). No genome-wide
             significant loci emerged for cannabis dependence criterion
             count (n=8050). There was also evidence that the minor
             allele of rs1409568 was associated with a 2.1% increase in
             right hippocampal volume in an independent sample of 430 EA
             college students (fwe-P=0.008). The identification and
             characterization of genome-wide significant loci for
             cannabis dependence is among the first steps toward
             understanding the biological contributions to the etiology
             of this psychiatric disorder, which appears to be rising in
             some developed nations.},
   Doi = {10.1038/mp.2017.200},
   Key = {fds335687}
}

@article{fds335690,
   Author = {Elliott, ML and Romer, A and Knodt, AR and Hariri,
             AR},
   Title = {A Connectome Wide Functional Signature of Transdiagnostic
             Risk for Mental Illness},
   Journal = {Biological Psychiatry},
   Volume = {84},
   Number = {6},
   Pages = {452-459},
   Year = {2018},
   Month = {April},
   Abstract = {Background High rates of comorbidity, shared risk, and
             overlapping therapeutic mechanisms have led psychopathology
             research towards transdiagnostic dimensional investigations
             of clustered symptoms. One influential framework accounts
             for these transdiagnostic phenomena through a single general
             factor, sometimes referred to as the ‘p’ factor,
             associated with risk for all common forms of mental illness.
             Methods Here we build on past research identifying unique
             structural neural correlates of the p factor by conducting a
             data-driven analysis of connectome wide intrinsic functional
             connectivity (n = 605). Results We demonstrate that higher p
             factor scores and associated risk for common mental illness
             maps onto hyper-connectivity between visual association
             cortex and both frontoparietal and default mode networks.
             Conclusions These results provide initial evidence that the
             transdiagnostic risk for common forms of mental illness is
             associated with patterns of inefficient connectome wide
             intrinsic connectivity between visual association cortex and
             networks supporting executive control and self-referential
             processes, networks which are often impaired across
             categorical disorders.},
   Doi = {10.1016/j.biopsych.2018.03.012},
   Key = {fds335690}
}

@article{fds335688,
   Author = {Chen, Q and Ursini, G and Romer, AL and Knodt, AR and Mezeivtch, K and Xiao, E and Pergola, G and Blasi, G and Straub, RE and Callicott, JH and Berman, KF and Hariri, AR and Bertolino, A and Mattay, VS and Weinberger, DR},
   Title = {Schizophrenia polygenic risk score predicts mnemonic
             hippocampal activity.},
   Journal = {Brain : a journal of neurology},
   Volume = {141},
   Number = {4},
   Pages = {1218-1228},
   Year = {2018},
   Month = {April},
   Abstract = {The use of polygenic risk scores has become a practical
             translational approach to investigating the complex genetic
             architecture of schizophrenia, but the link between
             polygenic risk scores and pathophysiological components of
             this disorder has been the subject of limited research. We
             investigated in healthy volunteers whether schizophrenia
             polygenic risk score predicts hippocampal activity during
             simple memory encoding, which has been proposed as a
             risk-associated intermediate phenotype of schizophrenia. We
             analysed the relationship between polygenic risk scores and
             hippocampal activity in a discovery sample of 191 unrelated
             healthy volunteers from the USA and in two independent
             replication samples of 76 and 137 healthy unrelated
             participants from Europe and the USA, respectively.
             Polygenic risk scores for each individual were calculated as
             the sum of the imputation probability of reference alleles
             weighted by the natural log of odds ratio from the recent
             schizophrenia genome-wide association study. We examined
             hippocampal activity during simple memory encoding of novel
             visual stimuli assessed using blood oxygen level-dependent
             functional MRI. Polygenic risk scores were significantly
             associated with hippocampal activity in the discovery sample
             [P = 0.016, family-wise error (FWE) corrected within
             Anatomical Automatic Labeling (AAL) bilateral
             hippocampal-parahippocampal mask] and in both replication
             samples (P = 0.033, FWE corrected within AAL right posterior
             hippocampal-parahippocampal mask in Bari sample, and P =
             0.002 uncorrected in the Duke Neurogenetics Study sample).
             The relationship between polygenic risk scores and
             hippocampal activity was consistently negative, i.e. lower
             hippocampal activity in individuals with higher polygenic
             risk scores, consistent with previous studies reporting
             decreased hippocampal-parahippocampal activity during
             declarative memory tasks in patients with schizophrenia and
             in their healthy siblings. Polygenic risk scores accounted
             for more than 8% of variance in hippocampal activity during
             memory encoding in discovery sample. We conclude that
             polygenic risk scores derived from the most recent
             schizophrenia genome-wide association study predict
             significant variability in hippocampal activity during
             memory encoding in healthy participants. Our findings
             validate mnemonic hippocampal activity as a genetic risk
             associated intermediate phenotype of schizophrenia,
             indicating that the aggregate neurobiological effect of
             schizophrenia risk alleles converges on this pattern of
             neural activity.awy004media15749593779001.},
   Doi = {10.1093/brain/awy004},
   Key = {fds335688}
}

@article{fds326208,
   Author = {Romer, AL and Knodt, AR and Houts, R and Brigidi, BD and Moffitt, TE and Caspi, A and Hariri, AR},
   Title = {Structural alterations within cerebellar circuitry are
             associated with general liability for common mental
             disorders.},
   Journal = {Molecular psychiatry},
   Volume = {23},
   Number = {4},
   Pages = {1084-1090},
   Year = {2018},
   Month = {April},
   Abstract = {Accumulating mental-health research encourages a shift in
             focus toward transdiagnostic dimensional features that are
             shared across categorical disorders. In support of this
             shift, recent studies have identified a general liability
             factor for psychopathology-sometimes called the 'p factor'-
             that underlies shared risk for a wide range of mental
             disorders. Identifying neural correlates of this general
             liability would substantiate its importance in
             characterizing the shared origins of mental disorders and
             help us begin to understand the mechanisms through which the
             'p factor' contributes to risk. Here we believe we first
             replicate the 'p factor' using cross-sectional data from a
             volunteer sample of 1246 university students, and then using
             high-resolution multimodal structural neuroimaging, we
             demonstrate that individuals with higher 'p factor' scores
             show reduced structural integrity of white matter pathways,
             as indexed by lower fractional anisotropy values, uniquely
             within the pons. Whole-brain analyses further revealed that
             higher 'p factor' scores are associated with reduced gray
             matter volume in the occipital lobe and left cerebellar
             lobule VIIb, which is functionally connected with prefrontal
             regions supporting cognitive control. Consistent with the
             preponderance of cerebellar afferents within the pons, we
             observed a significant positive correlation between the
             white matter integrity of the pons and cerebellar gray
             matter volume associated with higher 'p factor' scores. The
             results of our analyses provide initial evidence that
             structural alterations in corticocerebellar circuitry
             supporting core functions related to the basic integration,
             coordination and monitoring of information may contribute to
             a general liability for common mental disorders.},
   Doi = {10.1038/mp.2017.57},
   Key = {fds326208}
}

@article{fds332044,
   Author = {Scult, MA and Hariri, AR},
   Title = {A BRIEF INTRODUCTION TO THE NEUROGENETICS OF
             COGNITION-EMOTION INTERACTIONS.},
   Journal = {Current opinion in behavioral sciences},
   Volume = {19},
   Pages = {50-54},
   Year = {2018},
   Month = {February},
   Abstract = {Neuroscience research has demonstrated that cognition,
             emotion, and their dynamic interactions emerge from complex
             and flexible patterns of activity across distributed neural
             circuits. A parallel branch of research in genetics has
             begun to identify common variation in the human DNA sequence
             (i.e., genome) that may shape individual differences in
             cognition-emotion interactions by altering molecular and
             cellular pathways that modulate the activity of these neural
             circuits. Here we provide a brief introduction to such
             neurogenetics research and how it may usefully inform our
             understanding of the biological mechanisms through which
             dynamic cognition-emotion interactions emerge and,
             subsequently, help shape normal and abnormal
             behavior.},
   Doi = {10.1016/j.cobeha.2017.09.014},
   Key = {fds332044}
}

@article{fds331564,
   Author = {Swartz, JR and Knodt, AR and Radtke, SR and Hariri,
             AR},
   Title = {Post-secondary maternal education buffers against neural
             risk for psychological vulnerability to future life
             stress.},
   Journal = {Neuropsychologia},
   Volume = {109},
   Pages = {134-139},
   Publisher = {Elsevier BV},
   Year = {2018},
   Month = {January},
   Abstract = {We have previously reported that threat-related amygdala
             activity measured during a baseline fMRI scan predicts the
             experience of depression and anxiety associated with
             stressful life events years later. Here, we examine whether
             two broad measures of childhood environmental enrichment,
             namely parental educational achievement and subjective
             parental socioeconomic status, buffer against the effects of
             amygdala activity on future vulnerability to stress.
             Analyses of data available from 579 young adults revealed
             that maternal, but not paternal, educational achievement
             moderates the association between amygdala activity, recent
             life stress, and changes in mood and anxiety symptoms, even
             when controlling for participants' current subjective
             socioeconomic status. Specifically, only participants
             reporting lower maternal educational achievement exhibited
             our previously observed interaction between amygdala
             activity and future life stress predicting increases in
             depression and anxiety. These results suggest that higher
             maternal educational achievement may help buffer stress
             sensitivity associated with heightened threat-related
             amygdala activity.},
   Doi = {10.1016/j.neuropsychologia.2017.12.019},
   Key = {fds331564}
}

@article{fds332756,
   Author = {Nikolova, YS and Misquitta, KA and Rocco, BR and Prevot, TD and Knodt,
             AR and Ellegood, J and Voineskos, AN and Lerch, JP and Hariri, AR and Sibille, E and Banasr, M},
   Title = {Shifting priorities: highly conserved behavioral and brain
             network adaptations to chronic stress across
             species.},
   Journal = {Translational psychiatry},
   Volume = {8},
   Number = {1},
   Pages = {26},
   Year = {2018},
   Month = {January},
   Abstract = {Parallel clinical and preclinical research have begun to
             illuminate the biological basis of stress-related disorders,
             including major depression, but translational bridges
             informing discrete mechanistic targets for intervention are
             missing. To address this critical need, we used structural
             MRI in a mouse model and in a large human sample to examine
             stress effects on brain structure that may be conserved
             across species. Specifically, we focused on a previously
             unexplored approach, whole-brain structural covariance, as
             it reflects synchronized changes in neuroanatomy,
             potentially due to mutual trophic influences or shared
             plasticity across regions. Using the unpredictable chronic
             mild stress (UCMS) paradigm in mouse we first demonstrate
             that UCMS-induced elevated behavioral emotionality
             correlates with increased size of the amygdala and other
             corticolimbic regions. We further identify focal increases
             in the amygdala's 'hubness' (degree and strength) set
             against the background of a global stress-related loss of
             network clustering and modularity. These macroscopic changes
             are supported on the molecular level by increased
             postsynaptic density-95 protein in the amygdala, consistent
             with stress-induced plastic changes and synaptic
             strengthening. Finally, we provide clinical evidence that
             strikingly similar structural network reorganization
             patterns exist in young adults reporting high childhood
             trauma and increased mood symptoms. Collectively, we provide
             initial translational evidence for a conserved
             stress-related increase in amygdala-centered structural
             synchrony, as measured by enhanced structural covariance,
             which is paralleled by a decrease in global structural
             synchrony. This putative trade-off reflected in increased
             amygdala-centered plastic changes at the expense of global
             structural dedifferentiation may represent a mechanistic
             pathway for depression and related psychopathology.},
   Doi = {10.1038/s41398-017-0083-5},
   Key = {fds332756}
}

@article{fds364156,
   Author = {d'Arbeloff, TC and Freedy, KR and Knodt, AR and Radtke, SR and Brigidi,
             BD and Hariri, AR},
   Title = {Emotion Regulation and the Experience of Future Negative
             Mood: The Importance of Assessing Social
             Support.},
   Journal = {Frontiers in psychology},
   Volume = {9},
   Pages = {2287},
   Year = {2018},
   Month = {January},
   Abstract = {Emotion regulation refers to the use of various strategies,
             such as cognitive reappraisal and expressive suppression, to
             help manage our negative experiences, emotions, and
             thoughts. Although such emotion regulation often occurs
             within broader social dynamics and interactions, little is
             known about how social contexts interact with specific
             regulation strategies to shape the experience of negative
             emotions. Using data from 544 young adult university
             students, we provide initial evidence that habitual use of
             cognitive reappraisal is associated with lower future
             experience of depression and anxiety primarily through
             higher perceived social support (PSS). In contrast,
             expressive suppression is associated with higher future
             depression and anxiety primarily through lower PSS. These
             patterns are consistent with the importance of interpersonal
             influences on emotion regulation and suggest that assessment
             of social support can help elucidate the mechanisms of
             successfully regulating negative mood.},
   Doi = {10.3389/fpsyg.2018.02287},
   Key = {fds364156}
}

@article{fds329769,
   Author = {Dotterer, HL and Waller, R and Neumann, CS and Shaw, DS and Forbes, EE and Hariri, AR and Hyde, LW},
   Title = {Examining the Factor Structure of the Self-Report of
             Psychopathy Short-Form Across Four Young Adult
             Samples.},
   Journal = {Assessment},
   Volume = {24},
   Number = {8},
   Pages = {1062-1079},
   Year = {2017},
   Month = {December},
   Abstract = {Psychopathy refers to a range of complex behaviors and
             personality traits, including callousness and antisocial
             behavior, typically studied in criminal populations. Recent
             studies have used self-reports to examine psychopathic
             traits among noncriminal samples. The goal of the current
             study was to examine the underlying factor structure of the
             Self-Report of Psychopathy Scale-Short Form (SRP-SF) across
             complementary samples and examine the impact of gender on
             factor structure. We examined the structure of the SRP-SF
             among 2,554 young adults from three undergraduate samples
             and a high-risk young adult sample. Using confirmatory
             factor analysis, a four-correlated factor model and a
             four-bifactor model showed good fit to the data. Evidence of
             weak invariance was found for both models across gender.
             These findings highlight that the SRP-SF is a useful measure
             of low-level psychopathic traits in noncriminal samples,
             although the underlying factor structure may not fully
             translate across men and women.},
   Doi = {10.1177/1073191116640355},
   Key = {fds329769}
}

@article{fds330405,
   Author = {Lam, M and Trampush, JW and Yu, J and Knowles, E and Davies, G and Liewald,
             DC and Starr, JM and Djurovic, S and Melle, I and Sundet, K and Christoforou, A and Reinvang, I and DeRosse, P and Lundervold, AJ and Steen, VM and Espeseth, T and Räikkönen, K and Widen, E and Palotie,
             A and Eriksson, JG and Giegling, I and Konte, B and Roussos, P and Giakoumaki, S and Burdick, KE and Payton, A and Ollier, W and Chiba-Falek, O and Attix, DK and Need, AC and Cirulli, ET and Voineskos,
             AN and Stefanis, NC and Avramopoulos, D and Hatzimanolis, A and Arking,
             DE and Smyrnis, N and Bilder, RM and Freimer, NA and Cannon, TD and London,
             E and Poldrack, RA and Sabb, FW and Congdon, E and Conley, ED and Scult,
             MA and Dickinson, D and Straub, RE and Donohoe, G and Morris, D and Corvin,
             A and Gill, M and Hariri, AR and Weinberger, DR and Pendleton, N and Bitsios, P and Rujescu, D and Lahti, J and Le Hellard and S and Keller, MC and Andreassen, OA and Deary, IJ and Glahn, DC and Malhotra, AK and Lencz,
             T},
   Title = {Large-Scale Cognitive GWAS Meta-Analysis Reveals
             Tissue-Specific Neural Expression and Potential Nootropic
             Drug Targets.},
   Journal = {Cell Rep},
   Volume = {21},
   Number = {9},
   Pages = {2597-2613},
   Year = {2017},
   Month = {November},
   Abstract = {Here, we present a large (n = 107,207) genome-wide
             association study (GWAS) of general cognitive ability ("g"),
             further enhanced by combining results with a large-scale
             GWAS of educational attainment. We identified 70 independent
             genomic loci associated with general cognitive ability.
             Results showed significant enrichment for genes causing
             Mendelian disorders with an intellectual disability
             phenotype. Competitive pathway analysis implicated the
             biological processes of neurogenesis and synaptic
             regulation, as well as the gene targets of two pharmacologic
             agents: cinnarizine, a T-type calcium channel blocker, and
             LY97241, a potassium channel inhibitor. Transcriptome-wide
             and epigenome-wide analysis revealed that the implicated
             loci were enriched for genes expressed across all brain
             regions (most strongly in the cerebellum). Enrichment was
             exclusive to genes expressed in neurons but not
             oligodendrocytes or astrocytes. Finally, we report genetic
             correlations between cognitive ability and disparate
             phenotypes including psychiatric disorders, several
             autoimmune disorders, longevity, and maternal age at first
             birth.},
   Doi = {10.1016/j.celrep.2017.11.028},
   Key = {fds330405}
}

@article{fds343723,
   Author = {Scult, MA and Knodt, AR and Radtke, SR and Brigidi, BD and Hariri,
             AR},
   Title = {Prefrontal Executive Control Rescues Risk for Anxiety
             Associated with High Threat and Low Reward Brain
             Function},
   Journal = {Cerebral Cortex},
   Volume = {29},
   Number = {1},
   Pages = {70-76},
   Year = {2017},
   Month = {November},
   Abstract = {Compared with neural biomarkers of risk for mental illness,
             little is known about biomarkers of resilience. We explore
             if greater executive control-related prefrontal activity may
             function as a resilience biomarker by “rescuing” risk
             associated with higher threat-related amygdala and lower
             reward-related ventral striatum activity. Functional MRI was
             used to assay baseline threat-related amygdala,
             reward-related ventral striatum, and executive
             control-related prefrontal activity in 120 young adult
             volunteers. Participants provided self-reported mood and
             anxiety ratings at baseline and follow-up. A moderation
             model revealed a significant three-way interaction wherein
             higher amygdala and lower ventral striatum activity
             predicted increases in anxiety in those with average or low
             but not high prefrontal activity. This effect was specific
             to anxiety, with the neural biomarkers explaining ~10% of
             the variance in change over time, above and beyond baseline
             symptoms, sex, age, IQ, presence or absence of DMS-IV
             diagnosis, and both early and recent stress. Our findings
             are consistent with the importance of top-down executive
             control in adaptive regulation of negative emotions, and
             highlight a unique combination of neural biomarkers that may
             identify at-risk individuals for whom the adoption of
             strategies to improve executive control of negative emotions
             may prove particularly beneficial.},
   Doi = {10.1093/cercor/bhx304},
   Key = {fds343723}
}

@article{fds332981,
   Author = {Lissek, T and Adams, M and Adelman, J and Ahissar, E and Akaaboune, M and Akil, H and al'Absi, M and Arain, F and Arango-Lasprilla, JC and Atasoy,
             D and Avila, J and Badawi, A and Bading, H and Baig, AM and Baleriola, J and Belmonte, C and Bertocchi, I and Betz, H and Blakemore, C and Blanke, O and Boehm-Sturm, P and Bonhoeffer, T and Bonifazi, P and Brose, N and Campolongo, P and Celikel, T and Chang, CC and Chang, T-Y and Citri, A and Cline, HT and Cortes, JM and Cullen, K and Dean, K and Delgado-Garcia,
             JM and Desroches, M and Disterhoft, JF and Dowling, JE and Draguhn, A and El-Khamisy, SF and El Manira and A and Enam, SA and Encinas, JM and Erramuzpe, A and Esteban, JA and Fariñas, I and Fischer, E and Fukunaga, I and Gabilondo, I and Ganten, D and Gidon, A and Gomez-Esteban, JC and Greengard, P and Grinevich, V and Gruart, A and Guillemin, R and Hariri, AR and Hassan, B and Häusser, M and Hayashi,
             Y and Hussain, NK and Jabbar, AA and Jaber, M and Jahn, R and Janahi, EM and Kabbaj, M and Kettenmann, H and Kindt, M and Knafo, S and Köhr, G and Komai, S and Krugers, H and Kuhn, B and Ghazal, NL and Larkum, ME and London, M and Lutz, B and Matute, C and Martinez-Millan, L and Maroun,
             M and McGaugh, J and Moustafa, AA and Nasim, A and Nave, K-A and Neher, E and Nikolich, K and Outeiro, T and Palmer, LM and Penagarikano, O and Perez-Otano, I and Pfaff, DW and Poucet, B and Rahman, A-U and Ramos-Cabrer, P and Rashidy-Pour, A and Roberts, RJ and Rodrigues, S and Sanes, JR and Schaefer, AT and Segal, M and Segev, I and Shafqat, S and Siddiqui, NA and Soreq, H and Soriano-García, E and Spanagel, R and Sprengel, R and Stuart, G and Südhof, TC and Tønnesen, J and Treviño,
             M and Uthman, BM and Venter, JC and Verkhratsky, A and Weiss, C and Wiesel,
             TN and Yaksi, E and Yizhar, O and Young, LJ and Young, P and Zawia, NH and Zugaza, JL and Hasan, MT},
   Title = {Building Bridges through Science.},
   Journal = {Neuron},
   Volume = {96},
   Number = {4},
   Pages = {730-735},
   Year = {2017},
   Month = {November},
   Abstract = {Science is ideally suited to connect people from different
             cultures and thereby foster mutual understanding. To promote
             international life science collaboration, we have launched
             "The Science Bridge" initiative. Our current project focuses
             on partnership between Western and Middle Eastern
             neuroscience communities.},
   Doi = {10.1016/j.neuron.2017.09.028},
   Key = {fds332981}
}

@article{fds330404,
   Author = {Schaefer, JD and Scult, MA and Caspi, A and Arseneault, L and Belsky,
             DW and Hariri, AR and Harrington, H and Houts, R and Ramrakha, S and Poulton, R and Moffitt, TE},
   Title = {Is low cognitive functioning a predictor or consequence of
             major depressive disorder? A test in two longitudinal birth
             cohorts.},
   Journal = {Development and psychopathology},
   Pages = {1-15},
   Year = {2017},
   Month = {November},
   Abstract = {Cognitive impairment has been identified as an important
             aspect of major depressive disorder (MDD). We tested two
             theories regarding the association between MDD and cognitive
             functioning using data from longitudinal cohort studies. One
             theory, the cognitive reserve hypothesis, suggests that
             higher cognitive ability in childhood decreases risk of
             later MDD. The second, the scarring hypothesis, instead
             suggests that MDD leads to persistent cognitive deficits
             following disorder onset. We tested both theories in the
             Dunedin Study, a population-representative cohort followed
             from birth to midlife and assessed repeatedly for both
             cognitive functioning and psychopathology. We also used data
             from the Environmental Risk Longitudinal Twin Study to test
             whether childhood cognitive functioning predicts future MDD
             risk independent of family-wide and genetic risk using a
             discordant twin design. Contrary to both hypotheses, we
             found that childhood cognitive functioning did not predict
             future risk of MDD, nor did study members with a past
             history of MDD show evidence of greater cognitive decline
             unless MDD was accompanied by other comorbid psychiatric
             conditions. Our results thus suggest that low cognitive
             functioning is related to comorbidity, but is neither an
             antecedent nor an enduring consequence of MDD. Future
             research may benefit from considering cognitive deficits
             that occur during depressive episodes from a transdiagnostic
             perspective.},
   Doi = {10.1017/s095457941700164x},
   Key = {fds330404}
}

@article{fds328846,
   Author = {Gard, AM and Waller, R and Shaw, DS and Forbes, EE and Hariri, AR and Hyde,
             LW},
   Title = {The long reach of early adversity: Parenting, stress, and
             neural pathways to antisocial behavior in
             adulthood.},
   Journal = {Biological psychiatry. Cognitive neuroscience and
             neuroimaging},
   Volume = {2},
   Number = {7},
   Pages = {582-590},
   Publisher = {Elsevier BV},
   Year = {2017},
   Month = {October},
   Abstract = {<h4>Background</h4>Early life adversities including harsh
             parenting, maternal depression, neighborhood deprivation,
             and low family economic resources are more prevalent in
             low-income urban environments and are potent predictors of
             psychopathology, including, for boys, antisocial behavior
             (AB). However, little research has examined how these
             stressful experiences alter later neural function. Moreover,
             identifying genetic markers of greater susceptibility to
             adversity is critical to understanding biopsychosocial
             pathways from early adversity to later psychopathology.<h4>Methods</h4>Within
             a sample of 310 low-income boys followed from age 1.5 to 20,
             multimethod assessments of adversities were examined at age
             2 and age 12. At age 20, amygdala reactivity to emotional
             facial expressions was assessed using fMRI, and symptoms of
             Antisocial Personality Disorder were assessed via structured
             clinical interview. Genetic variability in cortisol
             signaling (<i>CRHR1</i>) was examined as a moderator of
             pathways to amygdala reactivity.<h4>Results</h4>Observed
             parenting and neighborhood deprivation at age 2 each
             uniquely predicted amygdala reactivity to emotional faces at
             age 20 over and above other adversities measured at multiple
             developmental periods. Harsher parenting and greater
             neighborhood deprivation in toddlerhood predicted
             clinically-significant symptoms of AB via less amygdala
             reactivity to fearful facial expressions and this pathway
             was moderated by genetic variation in <i>CRHR1</i>.<h4>Conclusions</h4>These
             results elucidate a pathway linking early adversity to less
             amygdala reactivity to social signals of interpersonal
             distress 18 years later, which in turn increased risk for
             serious AB. Moreover, these findings suggest a genetic
             marker of youth more susceptible to adversity.},
   Doi = {10.1016/j.bpsc.2017.06.005},
   Key = {fds328846}
}

@article{fds329468,
   Author = {Avinun, R and Nevo, A and Knodt, AR and Elliott, ML and Radtke, SR and Brigidi, BD and Hariri, AR},
   Title = {Reward-Related Ventral Striatum Activity Buffers against the
             Experience of Depressive Symptoms Associated with Sleep
             Disturbances.},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {37},
   Number = {40},
   Pages = {9724-9729},
   Year = {2017},
   Month = {October},
   Abstract = {Sleep disturbances represent one risk factor for depression.
             Reward-related brain function, particularly the activity of
             the ventral striatum (VS), has been identified as a
             potential buffer against stress-related depression. We were
             therefore interested in testing whether reward-related VS
             activity would moderate the effect of sleep disturbances on
             depression in a large cohort of young adults. Data were
             available from 1129 university students (mean age 19.71 ±
             1.25 years; 637 women) who completed a reward-related
             functional MRI task to assay VS activity and provided
             self-reports of sleep using the Pittsburgh Sleep Quality
             Index and symptoms of depression using a summation of the
             General Distress/Depression and Anhedonic Depression
             subscales of the Mood and Anxiety Symptoms
             Questionnaire-short form. Analyses revealed that as VS
             activity increased the association between sleep
             disturbances and depressive symptoms decreased. The
             interaction between sleep disturbances and VS activity was
             robust to the inclusion of sex, age, race/ethnicity, past or
             present clinical disorder, early and recent life stress, and
             anxiety symptoms, as well as the interactions between VS
             activity and early or recent life stress as covariates. We
             provide initial evidence that high reward-related VS
             activity may buffer against depressive symptoms associated
             with poor sleep. Our analyses help advance an emerging
             literature supporting the importance of individual
             differences in reward-related brain function as a potential
             biomarker of relative risk for depression.<b>SIGNIFICANCE
             STATEMENT</b> Sleep disturbances are a common risk factor
             for depression. An emerging literature suggests that
             reward-related activity of the ventral striatum (VS), a
             brain region critical for motivation and goal-directed
             behavior, may buffer against the effect of negative
             experiences on the development of depression. Using data
             from a large sample of 1129 university students we
             demonstrate that as reward-related VS activity increases,
             the link between sleep disturbances and depression
             decreases. This finding contributes to accumulating research
             demonstrating that reward-related brain function may be a
             useful biomarker of relative risk for depression in the
             context of negative experiences.},
   Doi = {10.1523/jneurosci.1734-17.2017},
   Key = {fds329468}
}

@article{fds329035,
   Author = {Kim, MJ and Avinun, R and Knodt, AR and Radtke, SR and Hariri,
             AR},
   Title = {Neurogenetic plasticity and sex influence the link between
             corticolimbic structural connectivity and trait
             anxiety.},
   Journal = {Scientific reports},
   Volume = {7},
   Number = {1},
   Pages = {10959},
   Year = {2017},
   Month = {September},
   Abstract = {Corticolimbic pathways connecting the amygdala and ventral
             prefrontal cortex (vPFC) are linked with trait anxiety, but
             it remains unclear what potential genetic moderators
             contribute to this association. We sought to address this by
             examining the inter-individual variability in
             neuroplasticity as modeled by a functional polymorphism
             (rs6265) in the human gene for brain derived neurotrophic
             factor (BDNF). Amygdala-vPFC pathway fractional anisotropy
             (FA) from 669 diffusion magnetic resonance images was used
             to examine associations with trait anxiety as a function of
             rs6265 genotype. We first replicated the inverse correlation
             between trait anxiety and amygdala-vPFC pathway FA in women.
             Furthermore, we found a moderating influence of rs6265
             genotype such that the association between trait anxiety and
             right amygdala-vPFC pathway FA was strongest in women
             carrying the Met allele, which is linked with decreased
             activity-dependent neuroplasticity. Results indicate that
             the microstructural integrity of pathways supporting
             communication between the amygdala and vPFC help shape the
             expression of trait anxiety in women, and that this
             association is further modulated by genetically driven
             variability in neuroplasticity.},
   Doi = {10.1038/s41598-017-11497-2},
   Key = {fds329035}
}

@article{fds331565,
   Author = {Baranger, DAA and Margolis, S and Hariri, AR and Bogdan,
             R},
   Title = {An earlier time of scan is associated with greater
             threat-related amygdala reactivity.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {12},
   Number = {8},
   Pages = {1272-1283},
   Year = {2017},
   Month = {August},
   Abstract = {Time-dependent variability in mood and anxiety suggest that
             related neural phenotypes, such as threat-related amygdala
             reactivity, may also follow a diurnal pattern. Here, using
             data from 1,043 young adult volunteers, we found that
             threat-related amygdala reactivity was negatively coupled
             with time of day, an effect which was stronger in the left
             hemisphere (β = -0.1083, p-fdr = 0.0012). This
             effect was moderated by subjective sleep quality
             (β = -0.0715, p-fdr = 0.0387); participants who
             reported average and poor sleep quality had relatively
             increased left amygdala reactivity in the morning.
             Bootstrapped simulations suggest that similar
             cross-sectional samples with at least 300 participants would
             be able to detect associations between amygdala reactivity
             and time of scan. In control analyses, we found no
             associations between time and V1 activation. Our results
             provide initial evidence that threat-related amygdala
             reactivity may vary diurnally, and that this effect is
             potentiated among individuals with average to low sleep
             quality. More broadly, our results suggest that considering
             time of scan in study design or modeling time of scan in
             analyses, as well as collecting additional measures of
             circadian variation, may be useful for understanding
             threat-related neural phenotypes and their associations with
             behavior, such as fear conditioning, mood and anxiety
             symptoms, and related phenotypes.},
   Doi = {10.1093/scan/nsx057},
   Key = {fds331565}
}

@article{fds325037,
   Author = {Bogdan, R and Salmeron, BJ and Carey, CE and Agrawal, A and Calhoun, VD and Garavan, H and Hariri, AR and Heinz, A and Hill, MN and Holmes, A and Kalin, NH and Goldman, D},
   Title = {Imaging Genetics and Genomics in Psychiatry: A Critical
             Review of Progress and Potential.},
   Journal = {Biological psychiatry},
   Volume = {82},
   Number = {3},
   Pages = {165-175},
   Year = {2017},
   Month = {August},
   Abstract = {Imaging genetics and genomics research has begun to provide
             insight into the molecular and genetic architecture of
             neural phenotypes and the neural mechanisms through which
             genetic risk for psychopathology may emerge. As it
             approaches its third decade, imaging genetics is confronted
             by many challenges, including the proliferation of studies
             using small sample sizes and diverse designs, limited
             replication, problems with harmonization of neural
             phenotypes for meta-analysis, unclear mechanisms, and
             evidence that effect sizes may be more modest than
             originally posited, with increasing evidence of
             polygenicity. These concerns have encouraged the field to
             grow in many new directions, including the development of
             consortia and large-scale data collection projects and the
             use of novel methods (e.g., polygenic approaches, machine
             learning) that enhance the quality of imaging genetic
             studies but also introduce new challenges. We critically
             review progress in imaging genetics and offer suggestions
             and highlight potential pitfalls of novel approaches.
             Ultimately, the strength of imaging genetics and genomics
             lies in their translational and integrative potential with
             other research approaches (e.g., nonhuman animal models,
             psychiatric genetics, pharmacologic challenge) to elucidate
             brain-based pathways that give rise to the vast individual
             differences in behavior as well as risk for
             psychopathology.},
   Doi = {10.1016/j.biopsych.2016.12.030},
   Key = {fds325037}
}

@article{fds315805,
   Author = {Scult, MA and Knodt, AR and Hanson, JL and Ryoo, M and Adcock, RA and Hariri, AR and Strauman, TJ},
   Title = {Individual differences in regulatory focus predict neural
             response to reward.},
   Journal = {Soc Neurosci},
   Volume = {12},
   Number = {4},
   Pages = {419-429},
   Year = {2017},
   Month = {August},
   ISSN = {1747-0919},
   Abstract = {Although goal pursuit is related to both functioning of the
             brain's reward circuits and psychological factors, the
             literatures surrounding these concepts have often been
             separate. Here, we use the psychological construct of
             regulatory focus to investigate individual differences in
             neural response to reward. Regulatory focus theory proposes
             two motivational orientations for personal goal pursuit: (1)
             promotion, associated with sensitivity to potential gain,
             and (2) prevention, associated with sensitivity to potential
             loss. The monetary incentive delay task was used to
             manipulate reward circuit function, along with instructional
             framing corresponding to promotion and prevention in a
             within-subject design. We observed that the more promotion
             oriented an individual was, the lower their ventral striatum
             response to gain cues. Follow-up analyses revealed that
             greater promotion orientation was associated with decreased
             ventral striatum response even to no-value cues, suggesting
             that promotion orientation may be associated with relatively
             hypoactive reward system function. The findings are also
             likely to represent an interaction between the cognitive and
             motivational characteristics of the promotion system with
             the task demands. Prevention orientation did not correlate
             with ventral striatum response to gain cues, supporting the
             discriminant validity of regulatory focus theory. The
             results highlight a dynamic association between individual
             differences in self-regulation and reward system
             function.},
   Doi = {10.1080/17470919.2016.1178170},
   Key = {fds315805}
}

@article{fds325971,
   Author = {Di Iorio and CR and Carey, CE and Michalski, LJ and Corral-Frias, NS and Conley, ED and Hariri, AR and Bogdan, R},
   Title = {Hypothalamic-pituitary-adrenal axis genetic variation and
             early stress moderates amygdala function.},
   Journal = {Psychoneuroendocrinology},
   Volume = {80},
   Pages = {170-178},
   Year = {2017},
   Month = {June},
   Abstract = {Early life stress may precipitate psychopathology, at least
             in part, by influencing amygdala function. Converging
             evidence across species suggests that links between
             childhood stress and amygdala function may be dependent upon
             hypothalamic-pituitary-adrenal (HPA) axis function. Using
             data from college-attending non-Hispanic European-Americans
             (n=308) who completed the Duke Neurogenetics Study, we
             examined whether early life stress (ELS) and HPA axis
             genetic variation interact to predict threat-related
             amygdala function as well as psychopathology symptoms. A
             biologically-informed multilocus profile score (BIMPS)
             captured HPA axis genetic variation (FKBP5 rs1360780, CRHR1
             rs110402; NR3C2 rs5522/rs4635799) previously associated with
             its function (higher BIMPS are reflective of higher HPA axis
             activity). BOLD fMRI data were acquired while participants
             completed an emotional face matching task. ELS and
             depression and anxiety symptoms were measured using the
             childhood trauma questionnaire and the mood and anxiety
             symptom questionnaire, respectively. The interaction between
             HPA axis BIMPS and ELS was associated with right amygdala
             reactivity to threat-related stimuli, after accounting for
             multiple testing (empirical-p=0.016). Among individuals with
             higher BIMPS (i.e., the upper 21.4%), ELS was positively
             coupled with threat-related amygdala reactivity, which was
             absent among those with average or low BIMPS. Further,
             higher BIMPS were associated with greater self-reported
             anxious arousal, though there was no evidence that amygdala
             function mediated this relationship. Polygenic variation
             linked to HPA axis function may moderate the effects of
             early life stress on threat-related amygdala function and
             confer risk for anxiety symptomatology. However, what, if
             any, neural mechanisms may mediate the relationship between
             HPA axis BIMPS and anxiety symptomatology remains
             unclear.},
   Doi = {10.1016/j.psyneuen.2017.03.016},
   Key = {fds325971}
}

@article{fds325036,
   Author = {Dotterer, HL and Hyde, LW and Swartz, JR and Hariri, AR and Williamson,
             DE},
   Title = {Amygdala reactivity predicts adolescent antisocial behavior
             but not callous-unemotional traits.},
   Journal = {Developmental cognitive neuroscience},
   Volume = {24},
   Pages = {84-92},
   Year = {2017},
   Month = {April},
   Abstract = {Recent neuroimaging studies have suggested divergent
             relationships between antisocial behavior (AB) and
             callous-unemotional (CU) traits and amygdala reactivity to
             fearful and angry facial expressions in adolescents.
             However, little work has examined if these findings extend
             to dimensional measures of behavior in ethnically diverse,
             non-clinical samples, or if participant sex, ethnicity,
             pubertal stage, and age moderate associations. We examined
             links between amygdala reactivity and dimensions of AB and
             CU traits in 220 Hispanic and non-Hispanic Caucasian
             adolescents (age 11-15; 49.5% female; 38.2% Hispanic), half
             of whom had a family history for depression and thus were at
             relatively elevated risk for late starting, emotionally
             dysregulated AB. We found that AB was significantly related
             to increased right amygdala reactivity to angry facial
             expressions independent of sex, ethnicity, pubertal stage,
             age, and familial risk status for depression. CU traits were
             not related to fear- or anger-related amygdala reactivity.
             The present study further demonstrates that AB is related to
             increased amygdala reactivity to interpersonal threat cues
             in adolescents, and that this relationship generalizes
             across sex, ethnicity, pubertal stage, age, and familial
             risk status for depression.},
   Doi = {10.1016/j.dcn.2017.02.008},
   Key = {fds325036}
}

@article{fds325035,
   Author = {Swartz, JR and Prather, AA and Hariri, AR},
   Title = {Threat-related amygdala activity is associated with
             peripheral CRP concentrations in men but not
             women.},
   Journal = {Psychoneuroendocrinology},
   Volume = {78},
   Pages = {93-96},
   Year = {2017},
   Month = {April},
   Abstract = {Increased levels of peripheral inflammatory markers,
             including C-Reactive Protein (CRP), are associated with
             increased risk for depression, anxiety, and suicidality. The
             brain mechanisms that may underlie the association between
             peripheral inflammation and internalizing problems remain to
             be determined. The present study examines associations
             between peripheral CRP concentrations and threat-related
             amygdala activity, a neural biomarker of depression and
             anxiety risk, in a sample of 172 young adult undergraduate
             students. Participants underwent functional MRI scanning
             while performing an emotional face matching task to obtain a
             measure of threat-related amygdala activity to angry and
             fearful faces; CRP concentrations were assayed from dried
             blood spots. Results indicated a significant interaction
             between CRP and sex: in men, but not women, higher CRP was
             associated with higher threat-related amygdala activity.
             These results add to the literature finding associations
             between systemic levels of inflammation and brain function
             and suggest that threat-related amygdala activity may serve
             as a potential pathway through which heightened chronic
             inflammation may increase risk for mood and anxiety
             problems.},
   Doi = {10.1016/j.psyneuen.2017.01.024},
   Key = {fds325035}
}

@article{fds323253,
   Author = {Carey, CE and Knodt, AR and Conley, ED and Hariri, AR and Bogdan,
             R},
   Title = {Reward-related ventral striatum activity links polygenic
             risk for attention-deficit/hyperactivity disorder to
             problematic alcohol use in young adulthood.},
   Journal = {Biological psychiatry. Cognitive neuroscience and
             neuroimaging},
   Volume = {2},
   Number = {2},
   Pages = {180-187},
   Publisher = {Elsevier BV},
   Year = {2017},
   Month = {March},
   Abstract = {<h4>Background</h4>Problematic alcohol use in adolescence
             and adulthood is a common and often debilitating correlate
             of childhood attention-deficit/hyperactivity disorder
             (ADHD). Converging evidence suggests that ADHD and
             problematic alcohol use share a common additive genetic
             basis, which may be mechanistically related to
             reward-related brain function. In the current study, we
             examined whether polygenic risk for childhood ADHD is linked
             to problematic alcohol use in young adulthood through
             alterations in reward-related activity of the ventral
             striatum, a neural hub supporting appetitive behaviors and
             reinforcement learning.<h4>Methods</h4>Genomic,
             neuroimaging, and self-report data were available for 404
             non-Hispanic European-American participants who completed
             the ongoing Duke Neurogenetics Study. Polygenic risk scores
             for childhood ADHD were calculated based on a genome-wide
             association study meta-analysis conducted by the Psychiatric
             Genomics Consortium and tested for association with
             reward-related ventral striatum activity, measured using a
             number-guessing functional magnetic resonance imaging
             paradigm, and self-reported problematic alcohol use. A
             mediational model tested whether ventral striatum activity
             indirectly links polygenic risk for ADHD to problematic
             alcohol use.<h4>Results</h4>Despite having no main effect on
             problematic alcohol use, polygenic risk for childhood ADHD
             was indirectly associated with problematic alcohol use
             through increased reward-related ventral striatum
             activity.<h4>Conclusions</h4>Individual differences in
             reward-related brain function may, at least in part,
             mechanistically link polygenic risk for childhood ADHD to
             problematic alcohol use.},
   Doi = {10.1016/j.bpsc.2016.10.003},
   Key = {fds323253}
}

@article{fds323689,
   Author = {Trampush, JW and Yang, MLZ and Yu, J and Knowles, E and Davies, G and Liewald, DC and Starr, JM and Djurovic, S and Melle, I and Sundet, K and Christoforou, A and Reinvang, I and DeRosse, P and Lundervold, AJ and Steen, VM and Espeseth, T and Räikkönen, K and Widen, E and Palotie,
             A and Eriksson, JG and Giegling, I and Konte, B and Roussos, P and Giakoumaki, S and Burdick, KE and Payton, A and Ollier, W and Horan, M and Chiba-Falek, O and Attix, DK and Need, AC and Cirulli, ET and Voineskos,
             AN and Stefanis, NC and Avramopoulos, D and Hatzimanolis, A and Arking,
             DE and Smyrnis, N and Bilder, RM and Freimer, NA and Cannon, TD and London,
             E and Poldrack, RA and Sabb, FW and Congdon, E and Conley, ED and Scult,
             MA and Dickinson, D and Straub, RE and Donohoe, G and Morris, D and Corvin,
             A and Gill, M and Hariri, AR and Weinberger, DR and Pendleton, N and Bitsios, P and Rujescu, D and Lahti, J and Le Hellard and S and Keller, MC and Andreassen, OA and Deary, IJ and Glahn, DC and Malhotra, AK and Lencz,
             T},
   Title = {GWAS meta-analysis reveals novel loci and genetic correlates
             for general cognitive function: a report from the COGENT
             consortium.},
   Journal = {Mol Psychiatry},
   Volume = {22},
   Number = {3},
   Pages = {336-345},
   Year = {2017},
   Month = {March},
   Abstract = {The complex nature of human cognition has resulted in
             cognitive genomics lagging behind many other fields in terms
             of gene discovery using genome-wide association study (GWAS)
             methods. In an attempt to overcome these barriers, the
             current study utilized GWAS meta-analysis to examine the
             association of common genetic variation (~8M
             single-nucleotide polymorphisms (SNP) with minor allele
             frequency ⩾1%) to general cognitive function in a sample
             of 35 298 healthy individuals of European ancestry across
             24 cohorts in the Cognitive Genomics Consortium (COGENT). In
             addition, we utilized individual SNP lookups and polygenic
             score analyses to identify genetic overlap with other
             relevant neurobehavioral phenotypes. Our primary GWAS
             meta-analysis identified two novel SNP loci (top SNPs:
             rs76114856 in the CENPO gene on chromosome 2 and rs6669072
             near LOC105378853 on chromosome 1) associated with cognitive
             performance at the genome-wide significance level (P<5 ×
             10-8). Gene-based analysis identified an additional three
             Bonferroni-corrected significant loci at chromosomes
             17q21.31, 17p13.1 and 1p13.3. Altogether, common variation
             across the genome resulted in a conservatively estimated SNP
             heritability of 21.5% (s.e.=0.01%) for general cognitive
             function. Integration with prior GWAS of cognitive
             performance and educational attainment yielded several
             additional significant loci. Finally, we found robust
             polygenic correlations between cognitive performance and
             educational attainment, several psychiatric disorders, birth
             length/weight and smoking behavior, as well as a novel
             genetic association to the personality trait of openness.
             These data provide new insight into the genetics of
             neurocognitive function with relevance to understanding the
             pathophysiology of neuropsychiatric illness.},
   Doi = {10.1038/mp.2016.244},
   Key = {fds323689}
}

@article{fds330406,
   Author = {Swartz, JR and Waller, R and Bogdan, R and Knodt, AR and Sabhlok, A and Hyde, LW and Hariri, AR},
   Title = {A Common Polymorphism in a Williams Syndrome Gene Predicts
             Amygdala Reactivity and Extraversion in Healthy
             Adults.},
   Journal = {Biological psychiatry},
   Volume = {81},
   Number = {3},
   Pages = {203-210},
   Year = {2017},
   Month = {February},
   Abstract = {<h4>Background</h4>Williams syndrome (WS), a genetic
             disorder resulting from hemizygous microdeletion of
             chromosome 7q11.23, has emerged as a model for identifying
             the genetic architecture of socioemotional behavior. Common
             polymorphisms in GTF2I, which is found within the WS
             microdeletion, have been associated with reduced social
             anxiety in the general population. Identifying neural
             phenotypes affected by these polymorphisms would help
             advance our understanding not only of this specific genetic
             association but also of the broader neurogenetic mechanisms
             of variability in socioemotional behavior.<h4>Methods</h4>Through
             an ongoing parent protocol, the Duke Neurogenetics Study, we
             measured threat-related amygdala reactivity to fearful and
             angry facial expressions using functional magnetic resonance
             imaging, assessed trait personality using the Revised NEO
             Personality Inventory, and imputed GTF2I rs13227433 from
             saliva-derived DNA using custom Illumina arrays.
             Participants included 808 non-Hispanic Caucasian, African
             American, and Asian university students.<h4>Results</h4>The
             GTF2I rs13227433 AA genotype, previously associated with
             lower social anxiety, predicted decreased threat-related
             amygdala reactivity. An indirect effect of GTF2I genotype on
             the warmth facet of extraversion was mediated by decreased
             threat-related amygdala reactivity in women but not
             men.<h4>Conclusions</h4>A common polymorphism in the WS gene
             GTF2I associated with reduced social anxiety predicts
             decreased threat-related amygdala reactivity, which mediates
             an association between genotype and increased warmth in
             women. These results are consistent with reduced
             threat-related amygdala reactivity in WS and suggest that
             common variation in GTF2I contributes to broader variability
             in socioemotional brain function and behavior, with
             implications for understanding the neurogenetic bases of WS
             as well as social anxiety.},
   Doi = {10.1016/j.biopsych.2015.12.007},
   Key = {fds330406}
}

@article{fds322497,
   Author = {Swartz, JR and Knodt, AR and Radtke, SR and Hariri,
             AR},
   Title = {Peering into the brain to predict behavior: Peer-reported,
             but not self-reported, conscientiousness links
             threat-related amygdala activity to future problem
             drinking.},
   Journal = {NeuroImage},
   Volume = {146},
   Pages = {894-903},
   Year = {2017},
   Month = {February},
   Abstract = {Personality traits such as conscientiousness as
             self-reported by individuals can help predict a range of
             outcomes, from job performance to longevity. Asking others
             to rate the personality of their acquaintances often
             provides even better predictive power than using
             self-report. Here, we examine whether peer-reported
             personality can provide a better link between brain
             function, namely threat-related amygdala activity, and
             future health-related behavior, namely problem drinking,
             than self-reported personality. Using data from a sample of
             377 young adult university students who were rated on five
             personality traits by peers, we find that higher
             threat-related amygdala activity to fearful facial
             expressions is associated with higher peer-reported, but not
             self-reported, conscientiousness. Moreover, higher
             peer-reported, but not self-reported, conscientiousness
             predicts lower future problem drinking more than one year
             later, an effect specific to men. Remarkably, relatively
             higher amygdala activity has an indirect effect on future
             drinking behavior in men, linked by peer-reported
             conscientiousness to lower future problem drinking. Our
             results provide initial evidence that the perceived
             conscientiousness of an individual by their peers uniquely
             reflects variability in a core neural mechanism supporting
             threat responsiveness. These novel patterns further suggest
             that incorporating peer-reported measures of personality
             into individual differences research can reveal novel
             predictive pathways of risk and protection for problem
             behaviors.},
   Doi = {10.1016/j.neuroimage.2016.10.003},
   Key = {fds322497}
}

@article{fds316880,
   Author = {Swartz, JR and Hariri, AR and Williamson, DE},
   Title = {An epigenetic mechanism links socioeconomic status to
             changes in depression-related brain function in high-risk
             adolescents.},
   Journal = {Molecular psychiatry},
   Volume = {22},
   Number = {2},
   Pages = {209-214},
   Year = {2017},
   Month = {February},
   ISSN = {1359-4184},
   Abstract = {Identifying biological mechanisms through which the
             experience of adversity emerges as individual risk for
             mental illness is an important step toward developing
             strategies for personalized treatment and, ultimately,
             prevention. Preclinical studies have identified epigenetic
             modification of gene expression as one such mechanism.
             Recent clinical studies have suggested that epigenetic
             modification, particularly methylation of gene regulatory
             regions, also acts to shape human brain function associated
             with risk for mental illness. However, it is not yet clear
             whether differential gene methylation as a function of
             adversity contributes to the emergence of individual risk
             for mental illness. Using prospective longitudinal
             epigenetic, neuroimaging and behavioral data from 132
             adolescents, we demonstrate that changes in gene methylation
             associated with lower socioeconomic status (SES) predict
             changes in risk-related brain function. Specifically, we
             find that lower SES during adolescence is associated with an
             increase in methylation of the proximal promoter of the
             serotonin transporter gene, which predicts greater increases
             in threat-related amygdala reactivity. We subsequently
             demonstrate that greater increases in amygdala reactivity
             moderate the association between a positive family history
             for depression and the later manifestation of depressive
             symptoms. These initial results suggest a specific
             biological mechanism through which adversity contributes to
             altered brain function, which in turn moderates the
             emergence of general liability as individual risk for mental
             illness. If replicated, this prospective pathway may
             represent a novel target biomarker for intervention and
             prevention among high-risk individuals.},
   Doi = {10.1038/mp.2016.82},
   Key = {fds316880}
}

@article{fds318715,
   Author = {Swartz, JR and Prather, AA and Di Iorio and CR and Bogdan, R and Hariri,
             AR},
   Title = {A Functional Interleukin-18 Haplotype Predicts Depression
             and Anxiety through Increased Threat-Related Amygdala
             Reactivity in Women but Not Men.},
   Journal = {Neuropsychopharmacology : official publication of the
             American College of Neuropsychopharmacology},
   Volume = {42},
   Number = {2},
   Pages = {419-426},
   Year = {2017},
   Month = {January},
   Abstract = {Common functional polymorphisms in the gene encoding
             interleukin-18 (IL18), a cytokine belonging to the IL-1
             superfamily that can induce synthesis of several other
             cytokines, have been associated with major depressive
             episodes following the experience of stressful life events.
             The neural mechanisms underlying these associations remain
             unexamined. Here we use an imaging genetics strategy to
             examine the effects of risk-related IL18 haplotypes
             comprising rs187238 and rs1946518 on threat-related amygdala
             reactivity and, through an indirect effect, stress-related
             symptoms of depression and anxiety in 448 non-Hispanic
             Caucasian university students. Analyses indicated that women
             but not men possessing an IL18 haplotype comprising both
             risk-related alleles evidenced increased threat-related left
             centromedial amygdala reactivity relative to other haplotype
             groups. Moreover, in women only, increased threat-related
             left centromedial amygdala reactivity predicted increased
             symptoms of depression and anxiety in individuals also
             reporting higher levels of life stress. Path analyses
             revealed a significant indirect effect of IL18 risk
             haplotype on symptoms of depression and anxiety through
             increased threat-related amygdala reactivity. These results
             suggest that a common functional IL18 haplotype associated
             with heightened proinflammatory responses confers
             susceptibility to stress-related depression and anxiety
             through effects on threat-related amygdala function, a risk
             pathway specific to women. If replicated, these patterns can
             inform the search for personalized interventions targeting
             neurobiological pathways of risk associated with
             inflammation.},
   Doi = {10.1038/npp.2016.129},
   Key = {fds318715}
}

@article{fds323776,
   Author = {Scult, MA and Knodt, AR and Swartz, JR and Brigidi, BD and Hariri,
             AR},
   Title = {Thinking and Feeling: Individual Differences in Habitual
             Emotion Regulation and Stress-Related Mood are Associated
             with Prefrontal Executive Control.},
   Journal = {Clinical psychological science : a journal of the
             Association for Psychological Science},
   Volume = {5},
   Number = {1},
   Pages = {150-157},
   Year = {2017},
   Month = {January},
   Abstract = {Calculating math problems from memory may seem unrelated to
             everyday processing of emotions, but they have more in
             common than one might think. Prior research highlights the
             importance of the dorsolateral prefrontal cortex (dlPFC) in
             executive control, intentional emotion regulation, and
             experience of dysfunctional mood and anxiety. While it has
             been hypothesized that emotion regulation may be related to
             'cold' (ie. not emotion-related) executive control, this
             assertion has not been tested. We address this gap by
             providing evidence that greater dlPFC activity during 'cold'
             executive control is associated with increased use of
             cognitive reappraisal to regulate emotions in everyday life.
             We then demonstrate that in the presence of increased life
             stress, increased dlPFC activity is associated with lower
             mood and anxiety symptoms and clinical diagnoses.
             Collectively, our results encourage ongoing efforts to
             understand prefrontal executive control as a possible
             intervention target for improving emotion regulation in mood
             and anxiety disorders.},
   Doi = {10.1177/2167702616654688},
   Key = {fds323776}
}

@article{fds318712,
   Author = {Scult, MA and Paulli, AR and Mazure, ES and Moffitt, TE and Hariri, AR and Strauman, TJ},
   Title = {The association between cognitive function and subsequent
             depression: a systematic review and meta-analysis.},
   Journal = {Psychol Med},
   Volume = {47},
   Number = {1},
   Pages = {1-17},
   Year = {2017},
   Month = {January},
   Abstract = {Despite a growing interest in understanding the cognitive
             deficits associated with major depressive disorder (MDD), it
             is largely unknown whether such deficits exist before
             disorder onset or how they might influence the severity of
             subsequent illness. The purpose of the present study was to
             conduct a systematic review and meta-analysis of
             longitudinal datasets to determine whether cognitive
             function acts as a predictor of later MDD diagnosis or
             change in depression symptoms. Eligible studies included
             longitudinal designs with baseline measures of cognitive
             functioning, and later unipolar MDD diagnosis or symptom
             assessment. The systematic review identified 29
             publications, representing 34 unique samples, and 121 749
             participants, that met the inclusion/exclusion criteria.
             Quantitative meta-analysis demonstrated that higher
             cognitive function was associated with decreased levels of
             subsequent depression (r = -0.088, 95% confidence interval.
             -0.121 to -0.054, p < 0.001). However, sensitivity analyses
             revealed that this association is likely driven by
             concurrent depression symptoms at the time of cognitive
             assessment. Our review and meta-analysis indicate that the
             association between lower cognitive function and later
             depression is confounded by the presence of contemporaneous
             depression symptoms at the time of cognitive assessment.
             Thus, cognitive deficits predicting MDD likely represent
             deleterious effects of subclinical depression symptoms on
             performance rather than premorbid risk factors for
             disorder.},
   Doi = {10.1017/S0033291716002075},
   Key = {fds318712}
}

@article{fds318709,
   Author = {Pornpattananangkul, N and Hariri, AR and Harada, T and Mano, Y and Komeda, H and Parrish, TB and Sadato, N and Iidaka, T and Chiao,
             JY},
   Title = {Cultural influences on neural basis of inhibitory
             control.},
   Journal = {NeuroImage},
   Volume = {139},
   Pages = {114-126},
   Year = {2016},
   Month = {October},
   Abstract = {Research on neural basis of inhibitory control has been
             extensively conducted in various parts of the world. It is
             often implicitly assumed that neural basis of inhibitory
             control is universally similar across cultures. Here, we
             investigated the extent to which culture modulated
             inhibitory-control brain activity at both cultural-group and
             cultural-value levels of analysis. During fMRI scanning,
             participants from different cultural groups (including
             Caucasian-Americans and Japanese-Americans living in the
             United States and native Japanese living in Japan) performed
             a Go/No-Go task. They also completed behavioral surveys
             assessing cultural values of behavioral consistency, or the
             extent to which one's behaviors in daily life are consistent
             across situations. Across participants, the Go/No-Go task
             elicited stronger neural activity in several
             inhibitory-control areas, such as the inferior frontal gyrus
             (IFG) and anterior cingulate cortex (ACC). Importantly, at
             the cultural-group level, we found variation in left IFG
             (L-IFG) activity that was explained by a cultural region
             where participants lived in (as opposed to race).
             Specifically, L-IFG activity was stronger for native
             Japanese compared to Caucasian- and Japanese-Americans,
             while there was no systematic difference in L-IFG activity
             between Japanese- and Caucasian-Americans. At the
             cultural-value level, we found that participants who valued
             being "themselves" across situations (i.e., having high
             endorsement of behavioral consistency) elicited stronger
             rostral ACC activity during the Go/No-Go task. Altogether,
             our findings provide novel insight into how culture
             modulates the neural basis of inhibitory
             control.},
   Doi = {10.1016/j.neuroimage.2016.05.061},
   Key = {fds318709}
}

@article{fds318710,
   Author = {Nikolova, YS and Swartz, JR and Hariri, AR},
   Title = {Can we identify meaningful epigenetic effects on human brain
             function and related risk for mental illness?},
   Journal = {Epigenomics},
   Volume = {8},
   Number = {10},
   Pages = {1307-1310},
   Year = {2016},
   Month = {October},
   Doi = {10.2217/epi-2016-0099},
   Key = {fds318710}
}

@article{fds318711,
   Author = {Kragel, PA and Knodt, AR and Hariri, AR and LaBar,
             KS},
   Title = {Decoding Spontaneous Emotional States in the Human
             Brain},
   Journal = {PLoS Biol},
   Volume = {14},
   Number = {9},
   Pages = {e2000106},
   Publisher = {Public Library of Science},
   Year = {2016},
   Month = {September},
   Abstract = {<title>Author Summary</title> <p>Functional brain imaging
             techniques provide a window into neural activity
             underpinning diverse cognitive processes, including visual
             perception, decision-making, and memory, among many others.
             By treating functional imaging data as a pattern-recognition
             problem, similar to face- or character-recognition,
             researchers have successfully identified patterns of brain
             activity that predict specific mental states; for example,
             the kind of an object being viewed. Moreover, these methods
             are capable of predicting mental states in the absence of
             external stimulation. For example, pattern-classifiers
             trained on brain responses to visual stimuli can
             successfully predict the contents of imagery during sleep.
             This research shows that internally mediated brain activity
             can be used to infer subjective mental states; however, it
             is not known whether more complex emotional mental states
             can be decoded from neuroimaging data in the absence of
             experimental manipulations. Here we show that brain-based
             models of specific emotions can detect individual
             differences in mood and emotional traits and are consistent
             with self-reports of emotional experience during
             intermittent periods of wakeful rest. These findings show
             that the brain dynamically fluctuates among multiple
             distinct emotional states at rest. More practically, the
             results suggest that brain-based models of emotion may help
             assess emotional status in clinical settings, particularly
             in individuals incapable of providing self-report of their
             own emotional experience.</p>},
   Doi = {10.1371/journal.pbio.2000106},
   Key = {fds318711}
}

@article{fds318713,
   Author = {Demers, CH and Drabant Conley and E and Bogdan, R and Hariri,
             AR},
   Title = {Interactions Between Anandamide and Corticotropin-Releasing
             Factor Signaling Modulate Human Amygdala Function and Risk
             for Anxiety Disorders: An Imaging Genetics Strategy for
             Modeling Molecular Interactions.},
   Journal = {Biological psychiatry},
   Volume = {80},
   Number = {5},
   Pages = {356-362},
   Year = {2016},
   Month = {September},
   Abstract = {<h4>Background</h4>Preclinical models reveal that
             stress-induced amygdala activity and impairment in fear
             extinction reflect reductions in anandamide driven by
             corticotropin-releasing factor receptor type 1 (CRF1)
             potentiation of the anandamide catabolic enzyme fatty acid
             amide hydrolase.<h4>Methods</h4>Here, we provide clinical
             translation for the importance of these molecular
             interactions using an imaging genetics strategy to examine
             whether interactions between genetic polymorphisms
             associated with differential anandamide (FAAH rs324420) and
             CRF1 (CRHR1 rs110402) signaling modulate amygdala function
             and anxiety disorder diagnosis.<h4>Results</h4>Analyses
             revealed that individuals with a genetic background
             predicting relatively high anandamide and CRF1 signaling
             exhibited blunted basolateral amygdala habituation, which
             further mediated increased risk for anxiety disorders among
             these same individuals.<h4>Conclusions</h4>The convergence
             of preclinical and clinical data suggests that interactions
             between anandamide and CRF1 represent a fundamental
             molecular mechanism regulating amygdala function and
             anxiety. Our results further highlight the potential of
             imaging genetics to powerfully translate complex preclinical
             findings to clinically meaningful human phenotypes.},
   Doi = {10.1016/j.biopsych.2015.12.021},
   Key = {fds318713}
}

@article{fds318714,
   Author = {Waller, R and Corral-Frías, NS and Vannucci, B and Bogdan, R and Knodt,
             AR and Hariri, AR and Hyde, LW},
   Title = {An oxytocin receptor polymorphism predicts amygdala
             reactivity and antisocial behavior in men.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {11},
   Number = {8},
   Pages = {1218-1226},
   Year = {2016},
   Month = {August},
   Abstract = {Variability in oxytocin (OXT) signaling is associated with
             individual differences in sex-specific social behavior
             across species. The effects of OXT signaling on social
             behavior are, in part, mediated through its modulation of
             amygdala function. Here, we use imaging genetics to examine
             sex-specific effects of three single-nucleotide
             polymorphisms in the human oxytocin receptor gene (OXTR;
             rs1042778, rs53576 and rs2254298) on threat-related amygdala
             reactivity and social behavior in 406 Caucasians. Analyses
             revealed that among men but not women, OXTR rs1042778 TT
             genotype was associated with increased right amygdala
             reactivity to angry facial expressions, which was uniquely
             related to higher levels of antisocial behavior among men.
             Moderated meditation analysis suggested a trending indirect
             effect of OXTR rs1042778 TT genotype on higher antisocial
             behavior via increased right amygdala reactivity to angry
             facial expressions in men. Our results provide evidence
             linking genetic variation in OXT signaling to individual
             differences in amygdala function. The results further
             suggest that these pathways may be uniquely important in
             shaping antisocial behavior in men.},
   Doi = {10.1093/scan/nsw042},
   Key = {fds318714}
}

@article{fds318716,
   Author = {Krystal, JH and Abi-Dargham, A and Akbarian, S and Arnsten, AFT and Barch, DM and Bearden, CE and Braff, DL and Brown, ES and Bullmore, ET and Carlezon, WA and Carter, CS and Cook, EH and Daskalakis, ZJ and DiLeone,
             RJ and Duman, RS and Grace, AA and Hariri, AR and Harrison, PJ and Hiroi,
             N and Kenny, PJ and Kleinman, JE and Krystal, AD and Lewis, DA and Lipska,
             BK and Marder, SR and Mason, GF and Mathalon, DH and McClung, CA and McDougle, CJ and McIntosh, AM and McMahon, FJ and Mirnics, K and Monteggia, LM and Narendran, R and Nestler, EJ and Neumeister, A and O'Donovan, MC and Öngür, D and Pariante, CM and Paulus, MP and Pearlson, G and Phillips, ML and Pine, DS and Pizzagalli, DA and Pletnikov, MV and Ragland, JD and Rapoport, JL and Ressler, KJ and Russo, SJ and Sanacora, G and Sawa, A and Schatzberg, AF and Shaham, Y and Shamay-Tsoory, SG and Sklar, P and State, MW and Stein, MB and Strakowski, SM and Taylor, SF and Turecki, G and Turetsky, BI and Weissman, MM and Zachariou, V and Zarate, CA and Zubieta,
             J-K},
   Title = {Constance E. Lieber, Theodore R. Stanley, and the Enduring
             Impact of Philanthropy on Psychiatry Research.},
   Journal = {Biol Psychiatry},
   Volume = {80},
   Number = {2},
   Pages = {84-86},
   Year = {2016},
   Month = {July},
   Doi = {10.1016/j.biopsych.2016.05.004},
   Key = {fds318716}
}

@article{fds316881,
   Author = {Corral-Frías, NS and Pizzagalli, DA and Carré, JM and Michalski,
             LJ and Nikolova, YS and Perlis, RH and Fagerness, J and Lee, MR and Conley,
             ED and Lancaster, TM and Haddad, S and Wolf, A and Smoller, JW and Hariri,
             AR and Bogdan, R},
   Title = {COMT Val(158) Met genotype is associated with reward
             learning: a replication study and meta-analysis.},
   Journal = {Genes, brain, and behavior},
   Volume = {15},
   Number = {5},
   Pages = {503-513},
   Year = {2016},
   Month = {June},
   ISSN = {1601-1848},
   Abstract = {Identifying mechanisms through which individual differences
             in reward learning emerge offers an opportunity to
             understand both a fundamental form of adaptive responding as
             well as etiological pathways through which aberrant reward
             learning may contribute to maladaptive behaviors and
             psychopathology. One candidate mechanism through which
             individual differences in reward learning may emerge is
             variability in dopaminergic reinforcement signaling. A
             common functional polymorphism within the catechol-O-methyl
             transferase gene (COMT; rs4680, Val(158) Met) has been
             linked to reward learning, where homozygosity for the Met
             allele (linked to heightened prefrontal dopamine function
             and decreased dopamine synthesis in the midbrain) has been
             associated with relatively increased reward learning. Here,
             we used a probabilistic reward learning task to asses
             response bias, a behavioral form of reward learning, across
             three separate samples that were combined for analyses (age:
             21.80 ± 3.95; n = 392; 268 female; European-American: n =
             208). We replicate prior reports that COMT rs4680 Met allele
             homozygosity is associated with increased reward learning in
             European-American participants (β = 0.20, t = 2.75, P <
             0.01; ΔR(2) = 0.04). Moreover, a meta-analysis of 4
             studies, including the current one, confirmed the
             association between COMT rs4680 genotype and reward learning
             (95% CI -0.11 to -0.03; z = 3.2; P < 0.01). These results
             suggest that variability in dopamine signaling associated
             with COMT rs4680 influences individual differences in reward
             which may potentially contribute to psychopathology
             characterized by reward dysfunction.},
   Doi = {10.1111/gbb.12296},
   Key = {fds316881}
}

@article{fds251943,
   Author = {Nelson, EC and Agrawal, A and Heath, AC and Bogdan, R and Sherva, R and Zhang, B and Al-Hasani, R and Bruchas, MR and Chou, Y-L and Demers, CH and Carey, CE and Conley, ED and Fakira, AK and Farrer, LA and Goate, A and Gordon, S and Henders, AK and Hesselbrock, V and Kapoor, M and Lynskey,
             MT and Madden, PAF and Moron, JA and Rice, JP and Saccone, NL and Schwab,
             SG and Shand, FL and Todorov, AA and Wallace, L and Wang, T and Wray, NR and Zhou, X and Degenhardt, L and Martin, NG and Hariri, AR and Kranzler,
             HR and Gelernter, J and Bierut, LJ and Clark, DJ and Montgomery,
             GW},
   Title = {Evidence of CNIH3 involvement in opioid dependence.},
   Journal = {Molecular psychiatry},
   Volume = {21},
   Number = {5},
   Pages = {608-614},
   Year = {2016},
   Month = {May},
   ISSN = {1359-4184},
   Abstract = {Opioid dependence, a severe addictive disorder and major
             societal problem, has been demonstrated to be moderately
             heritable. We conducted a genome-wide association study in
             Comorbidity and Trauma Study data comparing opioid-dependent
             daily injectors (N=1167) with opioid misusers who never
             progressed to daily injection (N=161). The strongest
             associations, observed for CNIH3 single-nucleotide
             polymorphisms (SNPs), were confirmed in two independent
             samples, the Yale-Penn genetic studies of opioid, cocaine
             and alcohol dependence and the Study of Addiction: Genetics
             and Environment, which both contain non-dependent opioid
             misusers and opioid-dependent individuals. Meta-analyses
             found five genome-wide significant CNIH3 SNPs. The A allele
             of rs10799590, the most highly associated SNP, was robustly
             protective (P=4.30E-9; odds ratio 0.64 (95% confidence
             interval 0.55-0.74)). Epigenetic annotation predicts that
             this SNP is functional in fetal brain. Neuroimaging data
             from the Duke Neurogenetics Study (N=312) provide evidence
             of this SNP's in vivo functionality; rs10799590 A allele
             carriers displayed significantly greater right amygdala
             habituation to threat-related facial expressions, a
             phenotype associated with resilience to psychopathology.
             Computational genetic analyses of physical dependence on
             morphine across 23 mouse strains yielded significant
             correlations for haplotypes in CNIH3 and functionally
             related genes. These convergent findings support CNIH3
             involvement in the pathophysiology of opioid dependence,
             complementing prior studies implicating the
             α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)
             glutamate system.},
   Doi = {10.1038/mp.2015.102},
   Key = {fds251943}
}

@article{fds330407,
   Author = {Victor, EC and Hariri, AR},
   Title = {A neuroscience perspective on sexual risk behavior in
             adolescence and emerging adulthood.},
   Journal = {Development and psychopathology},
   Volume = {28},
   Number = {2},
   Pages = {471-487},
   Year = {2016},
   Month = {May},
   Abstract = {Late adolescence and emerging adulthood (specifically ages
             15-24) represent a period of heightened sexual risk taking
             resulting in the greatest annual rates of sexually
             transmitted infections and unplanned pregnancies in the US
             population. Ongoing efforts to prevent such negative
             consequences are likely to benefit from a deepening of our
             understanding of biological mechanisms through which sexual
             risk taking emerges and biases decision making during this
             critical window. Here we present a neuroscience framework
             from which a mechanistic examination of sexual risk taking
             can be advanced. Specifically, we adapt the
             neurodevelopmental triadic model, which outlines how
             motivated behavior is governed by three systems: approach,
             avoidance, and regulation, to sexual decision making and
             subsequent risk behavior. We further propose a testable
             hypothesis of the triadic model, wherein relatively
             decreased threat-related amygdala reactivity and increased
             reward-related ventral striatum reactivity leads to sexual
             risk taking, which is particularly exaggerated during
             adolescence and young adulthood when there is an
             overexpression of dopaminergic neurons coupled with immature
             top-down prefrontal cortex regulation. We conclude by
             discussing how future research based on our adapted triadic
             model can inform ongoing efforts to improve intervention and
             prevention efforts.},
   Doi = {10.1017/s0954579415001042},
   Key = {fds330407}
}

@article{fds313398,
   Author = {Gorka, AX and LaBar, KS and Hariri, AR},
   Title = {Variability in emotional responsiveness and coping style
             during active avoidance as a window onto psychological
             vulnerability to stress.},
   Journal = {Physiology & behavior},
   Volume = {158},
   Pages = {90-99},
   Year = {2016},
   Month = {May},
   ISSN = {0031-9384},
   Abstract = {Individual differences in coping styles are associated with
             psychological vulnerability to stress. Recent animal
             research suggests that coping styles reflect trade-offs
             between proactive and reactive threat responses during
             active avoidance paradigms, with proactive responses
             associated with better stress tolerance. Based on these
             preclinical findings, we developed a novel instructed active
             avoidance paradigm to characterize patterns of proactive and
             reactive responses using behavioral, motoric, and autonomic
             measures in humans. Analyses revealed significant
             inter-individual variability not only in the magnitude of
             general emotional responsiveness but also the likelihood to
             specifically express proactive or reactive responses. In men
             but not women, individual differences in general emotional
             responsiveness were linked to increased trait anxiety while
             proactive coping style was linked to increased trait
             aggression. These patterns are consistent with preclinical
             findings and suggest that instructed active avoidance
             paradigms may be useful in assessing psychological
             vulnerability to stress using objective behavioral
             measures.},
   Doi = {10.1016/j.physbeh.2016.02.036},
   Key = {fds313398}
}

@article{fds251941,
   Author = {Nikolova, YS and Knodt, AR and Radtke, SR and Hariri,
             AR},
   Title = {Divergent responses of the amygdala and ventral striatum
             predict stress-related problem drinking in young adults:
             possible differential markers of affective and impulsive
             pathways of risk for alcohol use disorder.},
   Journal = {Molecular psychiatry},
   Volume = {21},
   Number = {3},
   Pages = {348-356},
   Year = {2016},
   Month = {March},
   ISSN = {1359-4184},
   Abstract = {Prior work suggests that there may be two distinct pathways
             of alcohol use disorder (AUD) risk: one associated with
             positive emotion enhancement and behavioral impulsivity, and
             another associated with negative emotion relief and coping.
             We sought to map these two pathways onto individual
             differences in neural reward and threat processing assessed
             using blood-oxygen-level-dependent functional magnetic
             resonance imaging in a sample of 759 undergraduate students
             (426 women, mean age 19.65±1.24 years) participating in the
             Duke Neurogenetics Study. We demonstrate that problem
             drinking is highest in the context of stress and in those
             with one of two distinct neural phenotypes: (1) a
             combination of relatively low reward-related activity of the
             ventral striatum (VS) and high threat-related reactivity of
             the amygdala; or (2) a combination of relatively high VS
             activity and low amygdala reactivity. In addition, we
             demonstrate that the relationship between stress and problem
             alcohol use is mediated by impulsivity, as reflected in
             monetary delay discounting rates, for those with high VS-low
             amygdala reactivity, and by anxious/depressive
             symptomatology for those with the opposite neural risk
             phenotype. Across both neural phenotypes, we found that
             greater divergence between VS and amygdala reactivity
             predicted greater risk for problem drinking. Finally, for
             those individuals with the low VS-high amygdala risk
             phenotype we found that stress not only predicted the
             presence of AUD diagnosis at the time of neuroimaging but
             also subsequent problem drinking reported 3 months following
             study completion. These results offer new insight into the
             neural basis of AUD risk and suggest novel biological
             targets for early individualized treatment or
             prevention.},
   Doi = {10.1038/mp.2015.85},
   Key = {fds251941}
}

@article{fds314439,
   Author = {Baranger, DAA and Ifrah, C and Prather, AA and Carey, CE and Corral-Frías, NS and Drabant Conley and E and Hariri, AR and Bogdan,
             R},
   Title = {PER1 rs3027172 Genotype Interacts with Early Life Stress to
             Predict Problematic Alcohol Use, but Not Reward-Related
             Ventral Striatum Activity.},
   Journal = {Frontiers in psychology},
   Volume = {7},
   Pages = {464},
   Year = {2016},
   Month = {January},
   Abstract = {Increasing evidence suggests that the circadian and stress
             regulatory systems contribute to alcohol use disorder (AUD)
             risk, which may partially arise through effects on
             reward-related neural function. The C allele of the PER1
             rs3027172 single nucleotide polymorphism (SNP) reduces PER1
             expression in cells incubated with cortisol and has been
             associated with increased risk for adult AUD and problematic
             drinking among adolescents exposed to high levels of
             familial psychosocial adversity. Using data from
             undergraduate students who completed the ongoing Duke
             Neurogenetics Study (DNS) (n = 665), we tested whether
             exposure to early life stress (ELS; Childhood Trauma
             Questionnaire) moderates the association between rs3027172
             genotype and later problematic alcohol use (Alcohol Use
             Disorders Identification Test) as well as ventral striatum
             (VS) reactivity to reward (card-guessing task while
             functional magnetic resonance imaging data were acquired).
             Initial analyses found that PER1 rs3027172 genotype
             interacted with ELS to predict both problematic drinking and
             VS reactivity; minor C allele carriers, who were also
             exposed to elevated ELS reported greater problematic
             drinking and exhibited greater ventral striatum reactivity
             to reward-related stimuli. When gene × covariate and
             environment × covariate interactions were controlled for,
             the interaction predicting problematic alcohol use remained
             significant (p < 0.05, corrected) while the interaction
             predicting VS reactivity was no longer significant. These
             results extend our understanding of relationships between
             PER1 genotype, ELS, and problematic alcohol use, and serve
             as a cautionary tale on the importance of controlling for
             potential confounders in studies of moderation including
             gene × environment interactions.},
   Doi = {10.3389/fpsyg.2016.00464},
   Key = {fds314439}
}

@article{fds330408,
   Author = {Bogdan, R and Pagliaccio, D and Baranger, DA and Hariri,
             AR},
   Title = {Genetic Moderation of Stress Effects on Corticolimbic
             Circuitry.},
   Journal = {Neuropsychopharmacology : official publication of the
             American College of Neuropsychopharmacology},
   Volume = {41},
   Number = {1},
   Pages = {275-296},
   Year = {2016},
   Month = {January},
   Abstract = {Stress exposure is associated with individual differences in
             corticolimbic structure and function that often mirror
             patterns observed in psychopathology. Gene x environment
             interaction research suggests that genetic variation
             moderates the impact of stress on risk for psychopathology.
             On the basis of these findings, imaging genetics, which
             attempts to link variability in DNA sequence and structure
             to neural phenotypes, has begun to incorporate measures of
             the environment. This research paradigm, known as imaging
             gene x environment interaction (iGxE), is beginning to
             contribute to our understanding of the neural mechanisms
             through which genetic variation and stress increase
             psychopathology risk. Although awaiting replication,
             evidence suggests that genetic variation within the
             canonical neuroendocrine stress hormone system, the
             hypothalamic-pituitary-adrenal axis, contributes to
             variability in stress-related corticolimbic structure and
             function, which, in turn, confers risk for psychopathology.
             For iGxE research to reach its full potential it will have
             to address many challenges, of which we discuss: (i) small
             effects, (ii) measuring the environment and neural
             phenotypes, (iii) the absence of detailed mechanisms, and
             (iv) incorporating development. By actively addressing these
             challenges, iGxE research is poised to help identify the
             neural mechanisms underlying genetic and environmental
             associations with psychopathology.},
   Doi = {10.1038/npp.2015.216},
   Key = {fds330408}
}

@article{fds365877,
   Author = {Hyde, LW and Shaw, DS and Murray, L and Gard, A and Hariri, AR and Forbes,
             EE},
   Title = {Dissecting the Role of Amygdala Reactivity in Antisocial
             Behavior in a Sample of Young, Low-Income, Urban
             Men},
   Journal = {Clinical Psychological Science},
   Volume = {4},
   Number = {3},
   Pages = {527-544},
   Year = {2016},
   Month = {January},
   Abstract = {Neuroimaging has suggested that amygdala reactivity to
             emotional facial expressions is associated with antisocial
             behavior (AB), particularly among those high on
             callous–unemotional (CU) traits. To investigate this
             association and potential moderators of this relationship,
             including task/stimuli effects, subregional anatomy of the
             amygdala, and participant race, we used fMRI in a sample of
             167 racially diverse 20-year-old men from low-income
             families. We found that AB, but not CU traits, was
             negatively related to amygdala reactivity to fearful faces.
             This result was specific to fearful faces and strongest in
             the centromedial subregion of the amygdala. Arrest record
             was positively related to basolateral amygdala reactivity to
             fearful and angry faces. Results were strongest among those
             identified as African American and not present in those
             identified as European American. Our findings suggest
             substantial complexity in the relationship between amygdala
             function and AB reflecting moderating effects of task
             stimulus, subregional anatomy, and race.},
   Doi = {10.1177/2167702615614511},
   Key = {fds365877}
}

@article{fds318717,
   Author = {Carey, CE and Agrawal, A and Zhang, B and Conley, ED and Degenhardt, L and Heath, AC and Li, D and Lynskey, MT and Martin, NG and Montgomery, GW and Wang, T and Bierut, LJ and Hariri, AR and Nelson, EC and Bogdan,
             R},
   Title = {Monoacylglycerol lipase (MGLL) polymorphism rs604300
             interacts with childhood adversity to predict cannabis
             dependence symptoms and amygdala habituation: Evidence from
             an endocannabinoid system-level analysis.},
   Journal = {Journal of abnormal psychology},
   Volume = {124},
   Number = {4},
   Pages = {860-877},
   Year = {2015},
   Month = {November},
   Abstract = {Despite evidence for heritable variation in cannabis
             involvement and the discovery of cannabinoid receptors and
             their endogenous ligands, no consistent patterns have
             emerged from candidate endocannabinoid (eCB) genetic
             association studies of cannabis involvement. Given
             interactions between eCB and stress systems and associations
             between childhood stress and cannabis involvement, it may be
             important to consider childhood adversity in the context of
             eCB-related genetic variation. We employed a system-level
             gene-based analysis of data from the Comorbidity and Trauma
             Study (N = 1,558) to examine whether genetic variation in
             six eCB genes (anabolism: DAGLA, DAGLB, NAPEPLD; catabolism:
             MGLL, FAAH; binding: CNR1; SNPs N = 65) and childhood sexual
             abuse (CSA) predict cannabis dependence symptoms.
             Significant interactions with CSA emerged for MGLL at the
             gene level (p = .009), and for rs604300 within MGLL (ΔR2 =
             .007, p < .001), the latter of which survived SNP-level
             Bonferroni correction and was significant in an additional
             sample with similar directional effects (N = 859; ΔR2 =
             .005, p = .026). Furthermore, in a third sample (N = 312),
             there was evidence that rs604300 genotype interacts with
             early life adversity to predict threat-related basolateral
             amygdala habituation, a neural phenotype linked to the eCB
             system and addiction (ΔR2 = .013, p = .047). Rs604300 may
             be related to epigenetic modulation of MGLL expression.
             These results are consistent with rodent models implicating
             2-arachidonoylglycerol (2-AG), an endogenous cannabinoid
             metabolized by the enzyme encoded by MGLL, in the etiology
             of stress adaptation related to cannabis dependence, but
             require further replication.},
   Doi = {10.1037/abn0000079},
   Key = {fds318717}
}

@article{fds251944,
   Author = {Hanson, JL and Hariri, AR and Williamson, DE},
   Title = {Blunted Ventral Striatum Development in Adolescence Reflects
             Emotional Neglect and Predicts Depressive
             Symptoms.},
   Journal = {Biological psychiatry},
   Volume = {78},
   Number = {9},
   Pages = {598-605},
   Year = {2015},
   Month = {November},
   ISSN = {0006-3223},
   Abstract = {<h4>Background</h4>Emotional neglect is associated with
             multiple negative outcomes, particularly increased risk for
             depression. Motivated by increasing evidence of
             reward-related ventral striatum (VS) dysfunction in
             depression, we investigated the role of developmental
             changes in VS activity on the emergence of depressive
             symptomatology as a function of emotional
             neglect.<h4>Methods</h4>We examined relationships between
             longitudinal neuroimaging of reward-related VS activity,
             assessments of mood, and measures of emotional neglect in
             106 participants first scanned between ages 11 to 15 and
             then 2 years later.<h4>Results</h4>We found that greater
             levels of emotional neglect were associated with blunted
             development of reward-related VS activity between the first
             and second assessments (as indexed by lower residualized
             change scores). Additionally, we found that decreases in
             this reward-related VS activity were related to greater
             depressive symptomatology and partially mediated the
             association between emotional neglect and subsequent
             depressive symptomatology.<h4>Conclusions</h4>Our results
             provide an important demonstration that blunted development
             of reward-related VS activity as a function of emotional
             neglect predicts the emergence of depressive symptoms in
             adolescents. Further, our results are consistent with
             emerging evidence for the importance of reward-related VS
             dysfunction in the etiology and pathophysiology of
             depression. These results are a first step toward developing
             the ability to predict, prevent, and treat stress-related
             psychopathology through the targeting of specific neural
             phenotypes.},
   Doi = {10.1016/j.biopsych.2015.05.010},
   Key = {fds251944}
}

@article{fds330409,
   Author = {Hanson, JL and Knodt, AR and Brigidi, BD and Hariri,
             AR},
   Title = {Lower structural integrity of the uncinate fasciculus is
             associated with a history of child maltreatment and future
             psychological vulnerability to stress.},
   Journal = {Development and psychopathology},
   Volume = {27},
   Number = {4 Pt 2},
   Pages = {1611-1619},
   Year = {2015},
   Month = {November},
   Abstract = {The experience of child maltreatment is a significant risk
             factor for the development of later internalizing disorders
             such as depression and anxiety. This risk is particularly
             heightened after exposure to additional, more
             contemporaneous stress. While behavioral evidence exists for
             such "stress sensitization," little is known about the
             mechanisms mediating such relationships, particularly within
             the brain. Here we report that the experience of child
             maltreatment independent of recent life stress, gender, and
             age is associated with reduced structural integrity of the
             uncinate fasciculus, a major white matter pathway between
             the amygdala and ventromedial prefrontal cortex, in young
             adults. We further demonstrate that individuals with lower
             uncinate fasciculus integrity at baseline who subsequently
             experience stressful life events report higher levels of
             internalizing symptomatology at follow-up. Our findings
             suggest a novel neurobiological mechanism linking child
             maltreatment with later internalizing symptoms, specifically
             altered structural connectivity within the brain's
             threat-detection and emotion-regulation circuitry.},
   Doi = {10.1017/s0954579415000978},
   Key = {fds330409}
}

@article{fds315132,
   Author = {Hanson, JL and Albert, WD and Iselin, AR and Carré, JM and Dodge, KA and Hariri, AR},
   Title = {Cumulative Stress In Childhood is Associated with Blunted
             Reward-Related Brain Activity In Adulthood},
   Journal = {Social Cognitive and Affective Neuroscience},
   Volume = {11},
   Number = {3},
   Pages = {405-412},
   Year = {2015},
   Month = {October},
   ISSN = {1749-5016},
   url = {http://hdl.handle.net/10161/10777 Duke open access
             repository},
   Abstract = {Early life stress (ELS) is strongly associated with negative
             outcomes in adulthood, including reduced motivation and
             increased negative mood. The mechanisms mediating these
             relations, however, are poorly understood. We examined the
             relation between exposure to ELS and reward-related brain
             activity, which is known to predict motivation and mood, at
             age 26, in a sample followed since kindergarten with annual
             assessments. Using functional neuroimaging, we assayed
             individual differences in the activity of the ventral
             striatum (VS) during the processing of monetary rewards
             associated with a simple card-guessing task, in a sample of
             72 male participants. We examined associations between a
             cumulative measure of ELS exposure and VS activity in
             adulthood. We found that greater levels of cumulative stress
             during childhood and adolescence predicted lower
             reward-related VS activity in adulthood. Extending this
             general developmental pattern, we found that exposure to
             stress early in development (between kindergarten and grade
             3) was significantly associated with variability in adult VS
             activity. Our results provide an important demonstration
             that cumulative life stress, especially during this
             childhood period, is associated with blunted reward-related
             VS activity in adulthood. These differences suggest
             neurobiological pathways through which a history of ELS may
             contribute to reduced motivation and increased negative
             mood.},
   Doi = {10.1093/scan/nsv124},
   Key = {fds315132}
}

@article{fds251937,
   Author = {Hariri, AR and Holmes, A},
   Title = {Finding translation in stress research.},
   Journal = {Nature neuroscience},
   Volume = {18},
   Number = {10},
   Pages = {1347-1352},
   Year = {2015},
   Month = {October},
   ISSN = {1097-6256},
   Abstract = {In our ongoing efforts to advance understanding of human
             diseases, translational research across rodents and humans
             on stress-related mental disorders stands out as a field
             that is producing discoveries that illuminate mechanisms of
             risk and pathophysiology at a brisk rate. Here we offer a
             Perspective on how a productive translational research
             dialog between preclinical models and clinical studies of
             these disorders is being powered by an ever-developing
             appreciation of the shared neural circuits and genetic
             architecture that moderate the response to stress across
             species. Working from these deep foundations, we discuss the
             approaches, both traditional and innovative, that have the
             potential to deliver a new generation of risk biomarkers and
             therapeutic strategies for stress-related
             disorders.},
   Doi = {10.1038/nn.4111},
   Key = {fds251937}
}

@article{fds291135,
   Author = {Scult, MA and Trampush, JW and Zheng, F and Conley, ED and Lencz, T and Malhotra, AK and Dickinson, D and Weinberger, DR and Hariri,
             AR},
   Title = {A Common Polymorphism in SCN2A Predicts General Cognitive
             Ability through Effects on PFC Physiology.},
   Journal = {Journal of cognitive neuroscience},
   Volume = {27},
   Number = {9},
   Pages = {1766-1774},
   Year = {2015},
   Month = {September},
   ISSN = {0898-929X},
   Abstract = {Here we provide novel convergent evidence across three
             independent cohorts of healthy adults (n = 531),
             demonstrating that a common polymorphism in the gene
             encoding the α2 subunit of neuronal voltage-gated type II
             sodium channels (SCN2A) predicts human general cognitive
             ability or "g." Using meta-analysis, we demonstrate that the
             minor T allele of a common polymorphism (rs10174400) in
             SCN2A is associated with significantly higher "g"
             independent of gender and age. We further demonstrate using
             resting-state fMRI data from our discovery cohort (n = 236)
             that this genetic advantage may be mediated by increased
             capacity for information processing between the dorsolateral
             PFC and dorsal ACC, which support higher cognitive
             functions. Collectively, these findings fill a gap in our
             understanding of the genetics of general cognitive ability
             and highlight a specific neural mechanism through which a
             common polymorphism shapes interindividual variation in
             "g."},
   Doi = {10.1162/jocn_a_00826},
   Key = {fds291135}
}

@article{fds251948,
   Author = {Nikolova, YS and Hariri, AR},
   Title = {Can we observe epigenetic effects on human brain
             function?},
   Journal = {Trends in cognitive sciences},
   Volume = {19},
   Number = {7},
   Pages = {366-373},
   Year = {2015},
   Month = {July},
   ISSN = {1364-6613},
   Abstract = {Imaging genetics has identified many contributions of DNA
             sequence variation to individual differences in brain
             function, behavior, and risk for psychopathology. Recent
             studies have extended this work beyond the genome by mapping
             epigenetic differences, specifically gene methylation in
             peripherally assessed DNA, onto variability in behaviorally
             and clinically relevant brain function. These data have
             generated understandable enthusiasm for the potential of
             such research to illuminate biological mechanisms of risk.
             We use our research on the effects of genetic and epigenetic
             variation in the human serotonin transporter on brain
             function to generate a guardedly optimistic opinion that the
             available data encourage continued research in this
             direction, and suggest strategies to promote faster
             progress.},
   Doi = {10.1016/j.tics.2015.05.003},
   Key = {fds251948}
}

@article{fds251949,
   Author = {Trampush, JW and Lencz, T and Knowles, E and Davies, G and Guha, S and Pe'er, I and Liewald, DC and Starr, JM and Djurovic, S and Melle, I and Sundet, K and Christoforou, A and Reinvang, I and Mukherjee, S and DeRosse, P and Lundervold, A and Steen, VM and John, M and Espeseth, T and Räikkönen, K and Widen, E and Palotie, A and Eriksson, JG and Giegling, I and Konte, B and Ikeda, M and Roussos, P and Giakoumaki, S and Burdick, KE and Payton, A and Ollier, W and Horan, M and Scult, M and Dickinson, D and Straub, RE and Donohoe, G and Morris, D and Corvin, A and Gill, M and Hariri, A and Weinberger, DR and Pendleton, N and Iwata, N and Darvasi, A and Bitsios, P and Rujescu, D and Lahti, J and Le Hellard and S and Keller, MC and Andreassen, OA and Deary, IJ and Glahn, DC and Malhotra,
             AK},
   Title = {Independent evidence for an association between general
             cognitive ability and a genetic locus for educational
             attainment.},
   Journal = {American journal of medical genetics. Part B,
             Neuropsychiatric genetics : the official publication of the
             International Society of Psychiatric Genetics},
   Volume = {168B},
   Number = {5},
   Pages = {363-373},
   Year = {2015},
   Month = {July},
   ISSN = {1552-4841},
   Abstract = {Cognitive deficits and reduced educational achievement are
             common in psychiatric illness; understanding the genetic
             basis of cognitive and educational deficits may be
             informative about the etiology of psychiatric disorders. A
             recent, large genome-wide association study (GWAS) reported
             a genome-wide significant locus for years of education,
             which subsequently demonstrated association to general
             cognitive ability ("g") in overlapping cohorts. The current
             study was designed to test whether GWAS hits for educational
             attainment are involved in general cognitive ability in an
             independent, large-scale collection of cohorts. Using
             cohorts in the Cognitive Genomics Consortium (COGENT; up to
             20,495 healthy individuals), we examined the relationship
             between g and variants associated with educational
             attainment. We next conducted meta-analyses with 24,189
             individuals with neurocognitive data from the educational
             attainment studies, and then with 53,188 largely independent
             individuals from a recent GWAS of cognition. A SNP
             (rs1906252) located at chromosome 6q16.1, previously
             associated with years of schooling, was significantly
             associated with g (P = 1.47 × 10(-4) ) in COGENT.
             The first joint analysis of 43,381 non-overlapping
             individuals for this a priori-designated locus was strongly
             significant (P = 4.94 × 10(-7) ), and the second
             joint analysis of 68,159 non-overlapping individuals was
             even more robust (P = 1.65 × 10(-9) ). These
             results provide independent replication, in a large-scale
             dataset, of a genetic locus associated with cognitive
             function and education. As sample sizes grow, cognitive GWAS
             will identify increasing numbers of associated loci, as has
             been accomplished in other polygenic quantitative traits,
             which may be relevant to psychiatric illness.},
   Doi = {10.1002/ajmg.b.32319},
   Key = {fds251949}
}

@article{fds251938,
   Author = {Telch, MJ and Beevers, CG and Rosenfield, D and Lee, H-J and Reijntjes,
             A and Ferrell, RE and Hariri, AR},
   Title = {5-HTTLPR genotype potentiates the effects of war zone
             stressors on the emergence of PTSD, depressive and anxiety
             symptoms in soldiers deployed to iraq.},
   Journal = {World psychiatry : official journal of the World Psychiatric
             Association (WPA)},
   Volume = {14},
   Number = {2},
   Pages = {198-206},
   Year = {2015},
   Month = {June},
   ISSN = {1723-8617},
   Abstract = {Exposure to war zone stressors is common, yet only a
             minority of soldiers experience clinically meaningful
             disturbance in psychological function. Identification of
             biomarkers that predict vulnerability to war zone stressors
             is critical for developing more effective treatment and
             prevention strategies not only in soldiers but also in
             civilians who are exposed to trauma. We investigated the
             role of the serotonin transporter linked polymorphic region
             (5-HTTLPR) genotype in predicting the emergence of
             post-traumatic stress disorder (PTSD), depressive and
             anxiety symptoms as a function of war zone stressors. A
             prospective cohort of 133 U.S. Army soldiers with no prior
             history of deployment to a war zone, who were scheduled to
             deploy to Iraq, was recruited. Multilevel regression models
             were used to investigate associations between 5-HTTLPR
             genotype, level of war zone stressors, and reported symptoms
             of PTSD, depression and anxiety while deployed to Iraq.
             Level of war zone stressors was associated with symptoms of
             PTSD, depression and anxiety. Consistent with its effects on
             stress responsiveness, 5-HTTLPR genotype moderated the
             relationship between level of war zone stressors and
             symptoms of emotional disturbance. Specifically, soldiers
             carrying one or two low functioning alleles (S or LG )
             reported heightened symptoms of PTSD, depression and anxiety
             in response to increased levels of exposure to war zone
             stressors, relative to soldiers homozygous for the high
             functioning allele (LA ). These data suggest that 5-HTTLPR
             genotype moderates individual sensitivity to war zone
             stressors and the expression of emotional disturbance
             including PTSD symptoms. Replication of this association
             along with identification of other genetic moderators of
             risk can inform the development of biomarkers that can
             predict relative resilience vs. vulnerability to
             stress.},
   Doi = {10.1002/wps.20215},
   Key = {fds251938}
}

@article{fds251939,
   Author = {Arloth, J and Bogdan, R and Weber, P and Frishman, G and Menke, A and Wagner, KV and Balsevich, G and Schmidt, MV and Karbalai, N and Czamara,
             D and Altmann, A and Trümbach, D and Wurst, W and Mehta, D and Uhr, M and Klengel, T and Erhardt, A and Carey, CE and Conley, ED and Major
             Depressive Disorder Working Group of the Psychiatric
             Genomics Consortium (PGC), and Ruepp, A and Müller-Myhsok, B and Hariri, AR and Binder, EB and Major Depressive Disorder Working
             Group of the Psychiatric Genomics Consortium
             PGC},
   Title = {Genetic Differences in the Immediate Transcriptome Response
             to Stress Predict Risk-Related Brain Function and
             Psychiatric Disorders.},
   Journal = {Neuron},
   Volume = {86},
   Number = {5},
   Pages = {1189-1202},
   Year = {2015},
   Month = {June},
   ISSN = {0896-6273},
   Abstract = {Depression risk is exacerbated by genetic factors and stress
             exposure; however, the biological mechanisms through which
             these factors interact to confer depression risk are poorly
             understood. One putative biological mechanism implicates
             variability in the ability of cortisol, released in response
             to stress, to trigger a cascade of adaptive genomic and
             non-genomic processes through glucocorticoid receptor (GR)
             activation. Here, we demonstrate that common genetic
             variants in long-range enhancer elements modulate the
             immediate transcriptional response to GR activation in human
             blood cells. These functional genetic variants increase risk
             for depression and co-heritable psychiatric disorders.
             Moreover, these risk variants are associated with
             inappropriate amygdala reactivity, a transdiagnostic
             psychiatric endophenotype and an important stress hormone
             response trigger. Network modeling and animal experiments
             suggest that these genetic differences in GR-induced
             transcriptional activation may mediate the risk for
             depression and other psychiatric disorders by altering a
             network of functionally related stress-sensitive genes in
             blood and brain.},
   Doi = {10.1016/j.neuron.2015.05.034},
   Key = {fds251939}
}

@article{fds251940,
   Author = {Victor, EC and Sansosti, AA and Bowman, HC and Hariri,
             AR},
   Title = {Differential patterns of amygdala and ventral striatum
             activation predict gender-specific changes in sexual risk
             behavior.},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {35},
   Number = {23},
   Pages = {8896-8900},
   Year = {2015},
   Month = {June},
   ISSN = {0270-6474},
   Abstract = {Although the initiation of sexual behavior is common among
             adolescents and young adults, some individuals express this
             behavior in a manner that significantly increases their risk
             for negative outcomes including sexually transmitted
             infections. Based on accumulating evidence, we have
             hypothesized that increased sexual risk behavior reflects,
             in part, an imbalance between neural circuits mediating
             approach and avoidance in particular as manifest by
             relatively increased ventral striatum (VS) activity and
             relatively decreased amygdala activity. Here, we test our
             hypothesis using data from seventy 18- to 22-year-old
             university students participating in the Duke Neurogenetics
             Study. We found a significant three-way interaction between
             amygdala activation, VS activation, and gender predicting
             changes in the number of sexual partners over time. Although
             relatively increased VS activation predicted greater
             increases in sexual partners for both men and women, the
             effect in men was contingent on the presence of relatively
             decreased amygdala activation and the effect in women was
             contingent on the presence of relatively increased amygdala
             activation. These findings suggest unique gender differences
             in how complex interactions between neural circuit function
             contributing to approach and avoidance may be expressed as
             sexual risk behavior in young adults. As such, our findings
             have the potential to inform the development of novel,
             gender-specific strategies that may be more effective at
             curtailing sexual risk behavior.},
   Doi = {10.1523/jneurosci.0737-15.2015},
   Key = {fds251940}
}

@article{fds251946,
   Author = {Gorka, AX and Norman, RE and Radtke, SR and Carré, JM and Hariri,
             AR},
   Title = {Anterior cingulate cortex gray matter volume mediates an
             association between 2D:4D ratio and trait aggression in
             women but not men.},
   Journal = {Psychoneuroendocrinology},
   Volume = {56},
   Pages = {148-156},
   Year = {2015},
   Month = {June},
   ISSN = {0306-4530},
   Abstract = {Previous research demonstrates that prenatal testosterone
             exposure increases aggression, possibly through its effects
             on the structure and function of neural circuits supporting
             threat detection and emotion regulation. Here we examined
             associations between regional gray matter volume, trait
             aggression, and the ratio of the second and fourth digit of
             the hand (2D:4D ratio) as a putative index of prenatal
             testosterone exposure in 464 healthy young adult volunteers.
             Our analyses revealed a significant positive correlation
             between 2D:4D ratio and gray matter volume of the dorsal
             anterior cingulate cortex (dACC), a brain region supporting
             emotion regulation, conflict monitoring, and behavioral
             inhibition. Subsequent analyses demonstrated that reduced
             (i.e., masculinized) gray matter volume in the dACC mediated
             the relationship between 2D:4D ratio and aggression in
             women, but not men. Expanding on this gender-specific
             mediation, additional analyses demonstrated that the shared
             variance between 2D:4D ratio, dACC gray matter volume, and
             aggression in women reflected the tendency to engage in
             cognitive reappraisal of emotionally provocative stimuli.
             Our results provide novel evidence that 2D:4D ratio is
             associated with masculinization of dACC gray matter volume,
             and that this neural phenotype mediates, in part, the
             expression of trait aggression in women.},
   Doi = {10.1016/j.psyneuen.2015.03.004},
   Key = {fds251946}
}

@article{fds251953,
   Author = {Kochunov, P and Jahanshad, N and Marcus, D and Winkler, A and Sprooten,
             E and Nichols, TE and Wright, SN and Hong, LE and Patel, B and Behrens, T and Jbabdi, S and Andersson, J and Lenglet, C and Yacoub, E and Moeller, S and Auerbach, E and Ugurbil, K and Sotiropoulos, SN and Brouwer, RM and Landman, B and Lemaitre, H and den Braber, A and Zwiers, MP and Ritchie,
             S and van Hulzen, K and Almasy, L and Curran, J and deZubicaray, GI and Duggirala, R and Fox, P and Martin, NG and McMahon, KL and Mitchell, B and Olvera, RL and Peterson, C and Starr, J and Sussmann, J and Wardlaw, J and Wright, M and Boomsma, DI and Kahn, R and de Geus, EJC and Williamson,
             DE and Hariri, A and van 't Ent, D and Bastin, ME and McIntosh, A and Deary, IJ and Hulshoff Pol and HE and Blangero, J and Thompson, PM and Glahn, DC and Van Essen and DC},
   Title = {Heritability of fractional anisotropy in human white matter:
             a comparison of Human Connectome Project and ENIGMA-DTI
             data.},
   Journal = {NeuroImage},
   Volume = {111},
   Pages = {300-311},
   Year = {2015},
   Month = {May},
   ISSN = {1053-8119},
   Abstract = {The degree to which genetic factors influence brain
             connectivity is beginning to be understood. Large-scale
             efforts are underway to map the profile of genetic effects
             in various brain regions. The NIH-funded Human Connectome
             Project (HCP) is providing data valuable for analyzing the
             degree of genetic influence underlying brain connectivity
             revealed by state-of-the-art neuroimaging methods. We
             calculated the heritability of the fractional anisotropy
             (FA) measure derived from diffusion tensor imaging (DTI)
             reconstruction in 481 HCP subjects (194/287 M/F) consisting
             of 57/60 pairs of mono- and dizygotic twins, and 246
             siblings. FA measurements were derived using (Enhancing
             NeuroImaging Genetics through Meta-Analysis) ENIGMA DTI
             protocols and heritability estimates were calculated using
             the SOLAR-Eclipse imaging genetic analysis package. We
             compared heritability estimates derived from HCP data to
             those publicly available through the ENIGMA-DTI consortium,
             which were pooled together from five-family based studies
             across the US, Europe, and Australia. FA measurements from
             the HCP cohort for eleven major white matter tracts were
             highly heritable (h(2)=0.53-0.90, p<10(-5)), and were
             significantly correlated with the joint-analytical estimates
             from the ENIGMA cohort on the tract and voxel-wise levels.
             The similarity in regional heritability suggests that the
             additive genetic contribution to white matter microstructure
             is consistent across populations and imaging acquisition
             parameters. It also suggests that the overarching genetic
             influence provides an opportunity to define a common genetic
             search space for future gene-discovery studies. Uniquely,
             the measurements of additive genetic contribution performed
             in this study can be repeated using online genetic analysis
             tools provided by the HCP ConnectomeDB web
             application.},
   Doi = {10.1016/j.neuroimage.2015.02.050},
   Key = {fds251953}
}

@article{fds251950,
   Author = {Gorka, AX and Knodt, AR and Hariri, AR},
   Title = {Basal forebrain moderates the magnitude of task-dependent
             amygdala functional connectivity.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {10},
   Number = {4},
   Pages = {501-507},
   Year = {2015},
   Month = {April},
   ISSN = {1749-5016},
   Abstract = {Animal studies reveal that the amygdala promotes attention
             and emotional memory, in part, by driving activity in
             downstream target regions including the prefrontal cortex
             (PFC) and hippocampus. Prior work has demonstrated that the
             amygdala influences these regions directly through
             monosynaptic glutamatergic signaling, and indirectly by
             driving activity of the cholinergic basal forebrain and
             subsequent downstream acetylcholine release. Yet to date, no
             work has addressed the functional relevance of the
             cholinergic basal forebrain in facilitating signaling from
             the amygdala in humans. We set out to determine how blood
             oxygen level-dependent signal within the amygdala and
             cholinergic basal forebrain interact to predict neural
             responses within downstream targets. Here, we use functional
             connectivity analyses to demonstrate that the cholinergic
             basal forebrain moderates increased amygdala connectivity
             with both the PFC and the hippocampus during the processing
             of biologically salient stimuli in humans. We further
             demonstrate that functional variation within the choline
             transporter gene predicts the magnitude of this modulatory
             effect. Collectively, our results provide novel evidence for
             the importance of cholinergic signaling in modulating neural
             pathways supporting arousal, attention and memory in humans.
             Further, our results may shed light on prior association
             studies linking functional variation within the choline
             transporter gene and diagnoses of major depression and
             attention-deficit hyperactivity disorder.},
   Doi = {10.1093/scan/nsu080},
   Key = {fds251950}
}

@article{fds251952,
   Author = {Swartz, JR and Williamson, DE and Hariri, AR},
   Title = {Developmental change in amygdala reactivity during
             adolescence: effects of family history of depression and
             stressful life events.},
   Journal = {The American journal of psychiatry},
   Volume = {172},
   Number = {3},
   Pages = {276-283},
   Year = {2015},
   Month = {March},
   ISSN = {0002-953X},
   Abstract = {<h4>Objective</h4>Although heightened amygdala reactivity is
             observed in patients with major depression, two critical
             gaps in our knowledge remain. First, it is unclear whether
             heightened amygdala reactivity is a premorbid vulnerability
             or a consequence of the disorder. Second, it is unknown how
             and when this neural phenotype develops. The authors sought
             to address these gaps by evaluating developmental change in
             threat-related amygdala reactivity in adolescents at high or
             low risk for depression based on family history, before
             onset of disorder.<h4>Method</h4>At baseline and again 2
             years later, adolescents (initially 11-15 years of age)
             participated in a functional MRI paradigm that elicited
             threat-related amygdala reactivity. After quality control,
             data were available for 232 adolescents at wave 1 and 197
             adolescents at wave 2; longitudinal data meeting quality
             control at both waves were available for 157 of these
             participants. Change in amygdala reactivity was assessed as
             a function of family history of depression and severity of
             stressful life events.<h4>Results</h4>Threat-related
             amygdala reactivity increased with age in participants with
             a positive family history regardless of the severity of life
             stress reported, and it increased in adolescents with a
             negative family history who reported relatively severe life
             stress. These changes in amygdala reactivity with age
             occurred in the absence of clinical disorder or increases in
             depressive symptoms.<h4>Conclusions</h4>These results
             suggest that heightened amygdala reactivity emerges during
             adolescence, prior to the development of depression, as a
             function of familial risk or, in the absence of familial
             risk, stressful life events.},
   Doi = {10.1176/appi.ajp.2014.14020195},
   Key = {fds251952}
}

@misc{fds251945,
   Author = {Faig, KE and Nikolova, YS and Hariri, AR},
   Title = {Genetic Neuroimaging of Social Perception},
   Volume = {3},
   Pages = {97-105},
   Booktitle = {Brain Mapping: An Encyclopedic Reference},
   Publisher = {Elsevier},
   Year = {2015},
   Month = {February},
   ISBN = {9780123973160},
   Doi = {10.1016/B978-0-12-397025-1.00161-5},
   Key = {fds251945}
}

@article{fds251955,
   Author = {Swartz, J and Knodt, A and Radtke, S and Hariri, A},
   Title = {A Neural Biomarker of Psychological Vulnerability to Future
             Life Stress},
   Journal = {Neuron},
   Volume = {85},
   Number = {3},
   Pages = {505-511},
   Publisher = {Elsevier},
   Year = {2015},
   Month = {February},
   ISSN = {0896-6273},
   url = {http://hdl.handle.net/10161/9483 Duke open access
             repository},
   Abstract = {We all experience a host of common life stressors such as
             the death of a family member, medical illness, and financial
             uncertainty. While most of us are resilient to such
             stressors, continuing to function normally, for a subset of
             individuals, experiencing these stressors increases the
             likelihood of developing treatment-resistant, chronic
             psychological problems, including depression and anxiety. It
             is thus paramount to identify predictive markers of risk,
             particularly those reflecting fundamental biological
             processes that can be targets for intervention and
             prevention. Using data from a longitudinal study of 340
             healthy young adults, we demonstrate that individual
             differences in threat-related amygdala reactivity predict
             psychological vulnerability to life stress occurring as much
             as 1 to 4 years later. These results highlight a readily
             assayed biomarker, threat-related amygdala reactivity, which
             predicts psychological vulnerability to commonly experienced
             stressors and represents a discrete target for intervention
             and prevention.},
   Doi = {10.1016/j.neuron.2014.12.055},
   Key = {fds251955}
}

@article{fds251951,
   Author = {Corral-Frías, NS and Nikolova, YS and Michalski, LJ and Baranger,
             DAA and Hariri, AR and Bogdan, R},
   Title = {Stress-related anhedonia is associated with ventral striatum
             reactivity to reward and transdiagnostic psychiatric
             symptomatology.},
   Journal = {Psychological medicine},
   Volume = {45},
   Number = {12},
   Pages = {2605-2617},
   Year = {2015},
   Month = {January},
   ISSN = {0033-2917},
   Abstract = {<h4>Background</h4>Early life stress (ELS) is consistently
             associated with increased risk for subsequent
             psychopathology. Individual differences in neural response
             to reward may confer vulnerability to stress-related
             psychopathology. Using data from the ongoing Duke
             Neurogenetics Study, the present study examined whether
             reward-related ventral striatum (VS) reactivity moderates
             the relationship between retrospectively reported ELS and
             anhedonic symptomatology. We further assessed whether
             individual differences in reward-related VS reactivity were
             associated with other depressive symptoms and problematic
             alcohol use via stress-related anhedonic symptoms and
             substance use-associated coping.<h4>Method</h4>Blood oxygen
             level-dependent functional magnetic resonance imaging (fMRI)
             was collected while participants (n = 906) completed a
             card-guessing task, which robustly elicits VS reactivity.
             ELS, anhedonic symptoms, other depressive symptoms, coping
             behavior, and alcohol use behavior were assessed with
             self-report questionnaires. Linear regressions were run to
             examine whether VS reactivity moderated the relationship
             between ELS and anhedonic symptoms. Structural equation
             models examined whether this moderation was indirectly
             associated with other depression symptoms and problematic
             alcohol use through its association with
             anhedonia.<h4>Results</h4>Analyses of data from 820
             participants passing quality control procedures revealed
             that the VS × ELS interaction was associated with anhedonic
             symptoms (p = 0.011). Moreover, structural equation models
             indirectly linked this interaction to non-anhedonic
             depression symptoms and problematic alcohol use through
             anhedonic symptoms and substance-related
             coping.<h4>Conclusions</h4>These findings suggest that
             reduced VS reactivity to reward is associated with increased
             risk for anhedonia in individuals exposed to ELS. Such
             stress-related anhedonia is further associated with other
             depressive symptoms and problematic alcohol use through
             substance-related coping.},
   Doi = {10.1017/s0033291715000525},
   Key = {fds251951}
}

@article{fds364221,
   Author = {Nikolova, YS and Iruku, SP and Lin, C-W and Conley, ED and Puralewski,
             R and French, B and Hariri, AR and Sibille, E},
   Title = {FRAS1-related extracellular matrix 3 (FREM3)
             single-nucleotide polymorphism effects on gene expression,
             amygdala reactivity and perceptual processing speed: An
             accelerated aging pathway of depression risk.},
   Journal = {Frontiers in psychology},
   Volume = {6},
   Pages = {1377},
   Year = {2015},
   Month = {January},
   Abstract = {The A allele of the FRAS1-related extracellular matrix
             protein 3 (FREM3) rs7676614 single nucleotide polymorphism
             (SNP) was linked to major depressive disorder (MDD) in an
             early genome-wide association study (GWAS), and to symptoms
             of psychomotor retardation in a follow-up investigation. In
             line with significant overlap between age- and
             depression-related molecular pathways, parallel work has
             shown that FREM3 expression in postmortem human brain
             decreases with age. Here, we probe the effect of rs7676614
             on amygdala reactivity and perceptual processing speed, both
             of which are altered in depression and aging. Amygdala
             reactivity was assessed using a face-matching BOLD fMRI
             paradigm in 365 Caucasian participants in the Duke
             Neurogenetics Study (DNS) (192 women, mean age 19.7 ± 1.2).
             Perceptual processing speed was indexed by reaction times in
             the same task and the Trail Making Test (TMT). The effect of
             rs7676614 on FREM3 mRNA brain expression levels was probed
             in a postmortem cohort of 169 Caucasian individuals (44
             women, mean age 50.8 ± 14.9). The A allele of rs7676614 was
             associated with blunted amygdala reactivity to faces, slower
             reaction times in the face-matching condition (p < 0.04), as
             well as marginally slower performance on TMT Part B (p =
             0.056). In the postmortem cohort, the T allele of rs6537170
             (proxy for the rs7676614 A allele), was associated with
             trend-level reductions in gene expression in Brodmann areas
             11 and 47 (p = 0.066), reminiscent of patterns
             characteristic of older age. The low-expressing allele of
             another FREM3 SNP (rs1391187) was similarly associated with
             reduced amygdala reactivity and slower TMT Part B speed, in
             addition to reduced BA47 activity and extraversion (p <
             0.05). Together, these results suggest common genetic
             variation associated with reduced FREM3 expression may
             confer risk for a subtype of depression characterized by
             reduced reactivity to environmental stimuli and slower
             perceptual processing speed, possibly suggestive of
             accelerated aging.},
   Doi = {10.3389/fpsyg.2015.01377},
   Key = {fds364221}
}

@article{fds251954,
   Author = {Carré, JM and Baird-Rowe, CD and Hariri, AR},
   Title = {Testosterone responses to competition predict decreased
             trust ratings of emotionally neutral faces.},
   Journal = {Psychoneuroendocrinology},
   Volume = {49},
   Pages = {79-83},
   Year = {2014},
   Month = {November},
   ISSN = {0306-4530},
   Abstract = {A wealth of evidence has linked individual differences in
             testosterone (T) to social, cognitive, and behavioral
             processes related to human dominance. Moreover, recent
             evidence indicates that a single administration of T reduces
             interpersonal trust in healthy young women. Here, in a
             sample of men and women (n=96), we investigated the extent
             to which endogenous fluctuations in T during a competitive
             interaction would predict subsequent ratings of trust from
             emotionally neutral faces. Results indicated that a rise in
             T predicted a decrease in trust ratings in men, but not
             women. These findings provide further support for the idea
             that competition-induced fluctuations in T may serve to
             modulate ongoing and/or future social behavior.},
   Doi = {10.1016/j.psyneuen.2014.06.011},
   Key = {fds251954}
}

@article{Ahs2013,
   Author = {Ahs, F and Davis, CF and Gorka, AX and Hariri, AR},
   Title = {Feature-based representations of emotional facial
             expressions in the human amygdala.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {9},
   Number = {9},
   Pages = {1372-1378},
   Address = {Center for Cognitive Neuroscience, Duke University Box
             90999, Durham, NC 27708, USA. fredrik.ahs@duke.edu.},
   Year = {2014},
   Month = {September},
   ISSN = {1749-5016},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23887817},
   Abstract = {The amygdala plays a central role in processing facial
             affect, responding to diverse expressions and features
             shared between expressions. Although speculation exists
             regarding the nature of relationships between expression-
             and feature-specific amygdala reactivity, this matter has
             not been fully explored. We used functional magnetic
             resonance imaging and principal component analysis (PCA) in
             a sample of 300 young adults, to investigate patterns
             related to expression- and feature-specific amygdala
             reactivity to faces displaying neutral, fearful, angry or
             surprised expressions. The PCA revealed a two-dimensional
             correlation structure that distinguished emotional
             categories. The first principal component separated neutral
             and surprised from fearful and angry expressions, whereas
             the second principal component separated neutral and angry
             from fearful and surprised expressions. This two-dimensional
             correlation structure of amygdala reactivity may represent
             specific feature-based cues conserved across discrete
             expressions. To delineate which feature-based cues
             characterized this pattern, face stimuli were averaged and
             then subtracted according to their principal component
             loadings. The first principal component corresponded to
             displacement of the eyebrows, whereas the second principal
             component corresponded to increased exposure of eye whites
             together with movement of the brow. Our results suggest a
             convergent representation of facial affect in the amygdala
             reflecting feature-based processing of discrete
             expressions.},
   Language = {Eng},
   Doi = {10.1093/scan/nst112},
   Key = {Ahs2013}
}

@article{fds251958,
   Author = {Nikolova, YS and Koenen, KC and Galea, S and Wang, C-M and Seney, ML and Sibille, E and Williamson, DE and Hariri, AR},
   Title = {Beyond genotype: serotonin transporter epigenetic
             modification predicts human brain function.},
   Journal = {Nature neuroscience},
   Volume = {17},
   Number = {9},
   Pages = {1153-1155},
   Year = {2014},
   Month = {September},
   ISSN = {1097-6256},
   Abstract = {We examined epigenetic regulation in regards to behaviorally
             and clinically relevant human brain function. Specifically,
             we found that increased promoter methylation of the
             serotonin transporter gene predicted increased
             threat-related amygdala reactivity and decreased mRNA
             expression in postmortem amygdala tissue. These patterns
             were independent of functional genetic variation in the same
             region. Furthermore, the association with amygdala
             reactivity was replicated in a second cohort and was robust
             to both sampling methods and age.},
   Doi = {10.1038/nn.3778},
   Key = {fds251958}
}

@article{fds251956,
   Author = {Bergman, O and Åhs, F and Furmark, T and Appel, L and Linnman, C and Faria, V and Bani, M and Pich, EM and Bettica, P and Henningsson, S and Manuck, SB and Ferrell, RE and Nikolova, YS and Hariri, AR and Fredrikson, M and Westberg, L and Eriksson, E},
   Title = {Association between amygdala reactivity and a dopamine
             transporter gene polymorphism.},
   Journal = {Translational psychiatry},
   Volume = {4},
   Pages = {e420},
   Year = {2014},
   Month = {August},
   Abstract = {Essential for detection of relevant external stimuli and for
             fear processing, the amygdala is under modulatory influence
             of dopamine (DA). The DA transporter (DAT) is of fundamental
             importance for the regulation of DA transmission by
             mediating reuptake inactivation of extracellular DA. This
             study examined if a common functional variable number tandem
             repeat polymorphism in the 3' untranslated region of the DAT
             gene (SLC6A3) influences amygdala function during the
             processing of aversive emotional stimuli. Amygdala
             reactivity was examined by comparing regional cerebral blood
             flow, measured with positron emission tomography and
             [(15)O]water, during exposure to angry and neutral faces,
             respectively, in a Swedish sample comprising 32 patients
             with social anxiety disorder and 17 healthy volunteers. In a
             separate US sample, comprising 85 healthy volunteers studied
             with blood oxygen level-dependent functional magnetic
             resonance imaging, amygdala reactivity was assessed by
             comparing the activity during exposure to threatening faces
             and neutral geometric shapes, respectively. In both the
             Swedish and the US sample, 9-repeat carriers displayed
             higher amygdala reactivity than 10-repeat homozygotes. The
             results suggest that this polymorphism contributes to
             individual variability in amygdala reactivity.},
   Doi = {10.1038/tp.2014.50},
   Key = {fds251956}
}

@article{fds251957,
   Author = {Goetz, SMM and Tang, L and Thomason, ME and Diamond, MP and Hariri, AR and Carré, JM},
   Title = {Testosterone rapidly increases neural reactivity to threat
             in healthy men: a novel two-step pharmacological challenge
             paradigm.},
   Journal = {Biological psychiatry},
   Volume = {76},
   Number = {4},
   Pages = {324-331},
   Year = {2014},
   Month = {August},
   ISSN = {0006-3223},
   Abstract = {<h4>Background</h4>Previous research suggests that
             testosterone (T) plays a key role in shaping competitive and
             aggressive behavior in humans, possibly by modulating
             threat-related neural circuitry. However, this research has
             been limited by the use of T augmentation that fails to
             account for baseline differences and has been conducted
             exclusively in women. Thus, the extent to which normal
             physiologic concentrations of T affect threat-related brain
             function in men remains unknown.<h4>Methods</h4>In the
             current study, we use a novel two-step pharmacologic
             challenge protocol to overcome these limitations and to
             evaluate causal modulation of threat- and aggression-related
             neural circuits by T in healthy young men (n = 16). First,
             we controlled for baseline differences in T through
             administration of a gonadotropin releasing hormone
             antagonist. Once a common baseline was established across
             participants, we then administered T to within the normal
             physiologic range. During this second step of the protocol
             we acquired functional neuroimaging data to examine the
             impact of T augmentation on neural circuitry supporting
             threat and aggression.<h4>Results</h4>Gonadotropin releasing
             hormone antagonism successfully reduced circulating
             concentrations of T and brought subjects to a common
             baseline. Administration of T rapidly increased circulating
             T concentrations and was associated with heightened
             reactivity of the amygdala, hypothalamus, and periaqueductal
             grey to angry facial expressions.<h4>Conclusions</h4>These
             findings provide novel causal evidence that T rapidly
             potentiates the response of neural circuits mediating threat
             processing and aggressive behavior in men.},
   Doi = {10.1016/j.biopsych.2014.01.016},
   Key = {fds251957}
}

@article{fds251959,
   Author = {Carré, JM and Iselin, A-MR and Welker, KM and Hariri, AR and Dodge,
             KA},
   Title = {Testosterone reactivity to provocation mediates the effect
             of early intervention on aggressive behavior.},
   Journal = {Psychological science},
   Volume = {25},
   Number = {5},
   Pages = {1140-1146},
   Year = {2014},
   Month = {May},
   ISSN = {0956-7976},
   Abstract = {We tested the hypotheses that the Fast Track intervention
             program for high-risk children would reduce adult aggressive
             behavior and that this effect would be mediated by decreased
             testosterone responses to social provocation. Participants
             were a subsample of males from the full trial sample, who
             during kindergarten had been randomly assigned to the
             10-year Fast Track intervention or to a control group. The
             Fast Track program attempted to develop children's social
             competencies through child social-cognitive and
             emotional-coping skills training, peer-relations coaching,
             academic tutoring, and classroom management, as well as
             training for parents to manage their child's behavior. At a
             mean age of 26 years, participants responded to laboratory
             provocations. Results indicated that, relative to control
             participants, men assigned to the intervention demonstrated
             reduced aggression and testosterone reactivity to social
             provocations. Moreover, reduced testosterone reactivity
             mediated the effect of intervention on aggressive behavior,
             which provides evidence for an enduring biological mechanism
             underlying the effect of early psychosocial intervention on
             aggressive behavior in adulthood.},
   Doi = {10.1177/0956797614525642},
   Key = {fds251959}
}

@article{fds303796,
   Author = {Gianaros, PJ and Marsland, AL and Kuan, DC-H and Schirda, BL and Jennings, JR and Sheu, LK and Hariri, AR and Gross, JJ and Manuck,
             SB},
   Title = {An inflammatory pathway links atherosclerotic cardiovascular
             disease risk to neural activity evoked by the cognitive
             regulation of emotion.},
   Journal = {Biological psychiatry},
   Volume = {75},
   Number = {9},
   Pages = {738-745},
   Year = {2014},
   Month = {May},
   ISSN = {0006-3223},
   Abstract = {<h4>Background</h4>Cognitive reappraisal is a form of
             emotion regulation that alters emotional responding by
             changing the meaning of emotional stimuli. Reappraisal
             engages regions of the prefrontal cortex that support
             multiple functions, including visceral control functions
             implicated in regulating the immune system. Immune activity
             plays a role in the preclinical pathophysiology of
             atherosclerotic cardiovascular disease (CVD), an
             inflammatory condition that is highly comorbid with
             affective disorders characterized by problems with emotion
             regulation. Here, we tested whether prefrontal engagement by
             reappraisal would be associated with atherosclerotic CVD
             risk and whether this association would be mediated by
             inflammatory activity.<h4>Methods</h4>Community volunteers
             (n = 157; 30-54 years of age; 80 women) without DSM-IV
             Axis-1 psychiatric diagnoses or cardiovascular or immune
             disorders performed a functional neuroimaging task involving
             the reappraisal of negative emotional stimuli. Carotid
             artery intima-media thickness and inter-adventitial diameter
             were measured by ultrasonography and used as markers of
             preclinical atherosclerosis. Also measured were circulating
             levels of interleukin-6 (IL-6), an inflammatory cytokine
             linked to CVD risk and prefrontal neural
             activity.<h4>Results</h4>Greater reappraisal-related
             engagement of the dorsal anterior cingulate cortex was
             associated with greater preclinical atherosclerosis and
             IL-6. Moreover, IL-6 mediated the association of dorsal
             anterior cingulate cortex engagement with preclinical
             atherosclerosis. These results were independent of age, sex,
             race, smoking status, and other known CVD risk
             factors.<h4>Conclusions</h4>The cognitive regulation of
             emotion might relate to CVD risk through a pathway involving
             the functional interplay between the anterior cingulate
             region of the prefrontal cortex and inflammatory
             activity.},
   Doi = {10.1016/j.biopsych.2013.10.012},
   Key = {fds303796}
}

@article{fds251963,
   Author = {McCarty, CA and Huggins, W and Aiello, AE and Bilder, RM and Hariri, A and Jernigan, TL and Newman, E and Sanghera, DK and Strauman, TJ and Zeng,
             Y and Ramos, EM and Junkins, HA and PhenX RISING network},
   Title = {PhenX RISING: real world implementation and sharing of PhenX
             measures.},
   Journal = {BMC Med Genomics},
   Volume = {7},
   Pages = {16},
   Year = {2014},
   Month = {March},
   Abstract = {BACKGROUND: The purpose of this manuscript is to describe
             the PhenX RISING network and the site experiences in the
             implementation of PhenX measures into ongoing
             population-based genomic studies. METHODS: Eighty PhenX
             measures were implemented across the seven PhenX RISING
             groups, thirty-three of which were used at more than two
             sites, allowing for cross-site collaboration. Each site used
             between four and 37 individual measures and five of the
             sites are validating the PhenX measures through comparison
             with other study measures. Self-administered and
             computer-based administration modes are being evaluated at
             several sites which required changes to the original PhenX
             Toolkit protocols. A network-wide data use agreement was
             developed to facilitate data sharing and collaboration.
             RESULTS: PhenX Toolkit measures have been collected for more
             than 17,000 participants across the PhenX RISING network.
             The process of implementation provided information that was
             used to improve the PhenX Toolkit. The Toolkit was revised
             to allow researchers to select self- or interviewer
             administration when creating the data collection worksheets
             and ranges of specimens necessary to run biological assays
             has been added to the Toolkit. CONCLUSIONS: The PhenX RISING
             network has demonstrated that the PhenX Toolkit measures can
             be implemented successfully in ongoing genomic studies. The
             next step will be to conduct gene/environment
             studies.},
   Doi = {10.1186/1755-8794-7-16},
   Key = {fds251963}
}

@article{fds251961,
   Author = {Hyde, LW and Byrd, AL and Votruba-Drzal, E and Hariri, AR and Manuck,
             SB},
   Title = {Amygdala reactivity and negative emotionality: divergent
             correlates of antisocial personality and psychopathy traits
             in a community sample.},
   Journal = {Journal of abnormal psychology},
   Volume = {123},
   Number = {1},
   Pages = {214-224},
   Year = {2014},
   Month = {February},
   ISSN = {0021-843X},
   Abstract = {Previous studies have emphasized that antisocial personality
             disorder (APD) and psychopathy overlap highly but differ
             critically in several features, notably negative
             emotionality (NEM) and possibly amygdala reactivity to
             social signals of threat and distress. Here we examined
             whether dimensions of psychopathy and APD correlate
             differentially with NEM and amygdala reactivity to emotional
             faces. Testing these relationships among healthy
             individuals, dimensions of psychopathy and APD were
             generated by the profile matching technique of Lynam and
             Widiger (2001), using facet scales of the NEO Personality
             Inventory-Revised, and amygdala reactivity was measured
             using a well-established emotional faces task, in a
             community sample of 103 men and women. Higher psychopathy
             scores were associated with lower NEM and lower amygdala
             reactivity, whereas higher APD scores were related to
             greater NEM and greater amygdala reactivity, but only after
             overlapping variance in APD and psychopathy was adjusted for
             in the statistical model. Amygdala reactivity did not
             mediate the relationship of APD and psychopathy scores to
             NEM. Supplemental analyses also compared other measures of
             factors within psychopathy in predicting NEM and amygdala
             reactivity and found that Factor 2 psychopathy was
             positively related to NEM and amygdala reactivity across
             measures of psychopathy. The overall findings replicate
             seminal observations on NEM in psychopathy by Hicks and
             Patrick (2006) and extend this work to neuroimaging in a
             normative population. They also suggest that one critical
             way in which APD and psychopathy dimensions may differ in
             their etiology is through their opposing levels of NEM and
             amygdala reactivity to threat.},
   Doi = {10.1037/a0035467},
   Key = {fds251961}
}

@article{fds251965,
   Author = {Roses, AD and Saunders, AM and Lutz, MW and Zhang, N and Hariri, AR and Asin, KE and Crenshaw, DG and Budur, K and Burns, DK and Brannan,
             SK},
   Title = {New applications of disease genetics and pharmacogenetics to
             drug development.},
   Journal = {Curr Opin Pharmacol},
   Volume = {14},
   Pages = {81-89},
   Year = {2014},
   Month = {February},
   ISSN = {1471-4892},
   Abstract = {TOMMORROW is a Phase III delay of onset clinical trial to
             determine whether low doses of pioglitazone, a molecule that
             induces mitochondrial doubling, delays the onset of MCI-AD
             in normal subjects treated with low dose compared to
             placebo. BOLD imaging studies in rodents and man were used
             to find the dose that increases oxygen consumption at
             central regions of the brain in higher proportion than
             activation of large corticol regions. The trial is made
             practical by the use of a pharmacogenetic algorithm based on
             TOMM40 and APOE genotypes and age to identify normal
             subjects at high risk of MCI-AD between the ages of 65-83
             years within a five year follow-up period.},
   Doi = {10.1016/j.coph.2013.12.002},
   Key = {fds251965}
}

@article{fds251967,
   Author = {Wellman, CL and Camp, M and Jones, VM and MacPherson, KP and Ihne, J and Fitzgerald, P and Maroun, M and Drabant, E and Bogdan, R and Hariri, AR and Holmes, A},
   Title = {Corrigendum to "Convergent effects of mouse Pet-1 deletion
             and human PET-1 variation on amygdala fear and threat
             processing" [Exp. Neurol. 250 (2013) 260-269]},
   Journal = {Experimental Neurology},
   Volume = {252},
   Pages = {104},
   Publisher = {Elsevier BV},
   Year = {2014},
   Month = {February},
   ISSN = {0014-4886},
   Doi = {10.1016/j.expneurol.2013.11.002},
   Key = {fds251967}
}

@article{fds251942,
   Author = {Gorka, AX and Hanson, JL and Radtke, SR and Hariri,
             AR},
   Title = {Reduced hippocampal and medial prefrontal gray matter
             mediate the association between reported childhood
             maltreatment and trait anxiety in adulthood and predict
             sensitivity to future life stress},
   Journal = {Biology of Mood and Anxiety Disorders},
   Volume = {4},
   Number = {1},
   Pages = {12-12},
   Publisher = {Springer Nature},
   Year = {2014},
   Month = {January},
   ISSN = {2045-5380},
   Abstract = {Background: The experience of early life stress is a
             consistently identified risk factor for the development of
             mood and anxiety disorders. Preclinical research employing
             animal models of early life stress has made inroads in
             understanding this association and suggests that the
             negative sequelae of early life stress may be mediated by
             developmental disruption of corticolimbic structures
             supporting stress responsiveness. Work in humans has
             corroborated this idea, as childhood adversity has been
             associated with alterations in gray matter volumes of the
             hippocampus, amygdala, and medial prefrontal cortex. Yet,
             missing from this body of research is a full understanding
             of how these neurobiological vulnerabilities may
             mechanistically contribute to the reported link between
             adverse childhood experiences and later affective
             psychopathology.Results: Analyses revealed that
             self-reported childhood maltreatment was associated with
             reduced gray matter volumes within the medial prefrontal
             cortex and left hippocampus. Furthermore, reduced left
             hippocampal and medial prefrontal gray matter volume
             mediated the relationship between childhood maltreatment and
             trait anxiety. Additionally, individual differences in
             corticolimbic gray matter volume within these same
             structures predicted the anxious symptoms as a function of
             life stress 1 year after initial assessment.Conclusions:
             Collectively, these findings provide novel evidence that
             reductions in corticolimbic gray matter, particularly within
             the hippocampus and medial prefrontal cortex, are associated
             with reported childhood maltreatment and individual
             differences in adult trait anxiety. Furthermore, our results
             suggest that these structural alterations contribute to
             increased affective sensitivity to stress later in life in
             those that have experienced early adversity. More broadly,
             the findings contribute to an emerging literature
             highlighting the critical importance of early stress on the
             development of corticolimbic structures supporting adaptive
             functioning later in life.},
   Doi = {10.1186/2045-5380-4-12},
   Key = {fds251942}
}

@article{Lohoff2013,
   Author = {Lohoff, FW and Hodge, R and Narasimhan, S and Nall, A and Ferraro, TN and Mickey, BJ and Heitzeg, MM and Langenecker, SA and Zubieta, J-K and Bogdan, R and Nikolova, YS and Drabant, E and Hariri, AR and Bevilacqua,
             L and Goldman, D and Doyle, GA},
   Title = {Functional genetic variants in the vesicular monoamine
             transporter 1 modulate emotion processing.},
   Journal = {Molecular psychiatry},
   Volume = {19},
   Number = {1},
   Pages = {129-139},
   Address = {Translational Research Laboratories, Department of
             Psychiatry, Center for Neurobiology and Behavior, University
             of Pennsylvania School of Medicine, Philadelphia, PA,
             USA.},
   Year = {2014},
   Month = {January},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23337945},
   Abstract = {Emotional behavior is in part heritable and often disrupted
             in psychopathology. Identification of specific genetic
             variants that drive this heritability may provide important
             new insight into molecular and neurobiological mechanisms
             involved in emotionality. Our results demonstrate that the
             presynaptic vesicular monoamine transporter 1 (VMAT1)
             Thr136Ile (rs1390938) polymorphism is functional in vitro,
             with the Ile allele leading to increased monoamine transport
             into presynaptic vesicles. Moreover, we show that the
             Thr136Ile variant predicts differential responses in
             emotional brain circuits consistent with its effects in
             vitro. Lastly, deep sequencing of bipolar disorder (BPD)
             patients and controls identified several rare novel VMAT1
             variants. The variant Phe84Ser was only present in
             individuals with BPD and leads to marked increase monoamine
             transport in vitro. Taken together, our data show that VMAT1
             polymorphisms influence monoamine signaling, the functional
             response of emotional brain circuits and risk for
             psychopathology.},
   Language = {Eng},
   Doi = {10.1038/mp.2012.193},
   Key = {Lohoff2013}
}

@article{fds251964,
   Author = {Goetz, SMM and Tang, L and Thomason, ME and Diamond, MP and Hariri, AR and Carré, JM},
   Title = {Testosterone Rapidly Increases Neural Reactivity to Threat
             in Healthy Men: A Novel Two-Step Pharmacological Challenge
             Paradigm},
   Journal = {Biological Psychiatry},
   Volume = {76},
   Number = {4},
   Pages = {324-331},
   Publisher = {Elsevier BV},
   Year = {2014},
   ISSN = {0006-3223},
   Doi = {10.1016/j.biopsych.2014.01.016},
   Key = {fds251964}
}

@article{Wellman2013,
   Author = {Wellman, CL and Camp, M and Jones, VM and MacPherson, KP and Ihne, J and Fitzgerald, P and Maroun, M and Drabant, E and Bogdan, R and Hariri, AR and Holmes, A},
   Title = {Convergent effects of mouse Pet-1 deletion and human PET-1
             variation on amygdala fear and threat processing.},
   Journal = {Experimental neurology},
   Volume = {250},
   Pages = {260-269},
   Address = {Department of Psychological and Brain Sciences, Center for
             the Integrative Study of Animal Behavior, Indiana
             University, Bloomington, IN, USA. Electronic address:
             wellmanc@indiana.edu.},
   Year = {2013},
   Month = {December},
   ISSN = {0014-4886},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/24100022},
   Abstract = {Serotonin is critical for shaping the development of neural
             circuits regulating emotion. Pet-1 (FEV-1) is an ETS-domain
             transcription factor essential for differentiation and
             forebrain targeting of serotonin neurons. Constitutive Pet-1
             knockout (KO) causes major loss of serotonin neurons and
             forebrain serotonin availability, and behavioral
             abnormalities. We phenotyped Pet-1 KO mice for fear
             conditioning and extinction, and on a battery of assays for
             anxiety- and depression-related behaviors. Morphology of
             Golgi-stained neurons in basolateral amygdala (BLA) and
             prelimbic cortex was examined. Using human imaging genetics,
             a common variant (rs860573) in the PET-1 (FEV) gene was
             tested for effects on threat-related amygdala reactivity and
             psychopathology in 88 Asian-ancestry subjects. Pet-1 KO mice
             exhibited increased acquisition and expression of fear, and
             elevated fear recovery following extinction, relative to
             wild-type (WT). BLA dendrites of Pet-1 KO mice were
             significantly longer than in WT. Human PET-1 variation
             associated with differences in amygdala threat processing
             and psychopathology. This novel evidence for the role of
             Pet-1 in fear processing and dendritic organization of
             amygdala neurons and in human amygdala threat processing
             extends a growing literature demonstrating the influence of
             genetic variation in the serotonin system on emotional
             regulation via effects on structure and function of
             underlying corticolimbic circuitry.},
   Language = {eng},
   Doi = {10.1016/j.expneurol.2013.09.025},
   Key = {Wellman2013}
}

@article{fds251968,
   Author = {Disner, SG and Beevers, CG and Lee, HJ and Ferrell, RE and Hariri, AR and Telch, MJ},
   Title = {War zone stress interacts with the 5-HTTLPR polymorphism to
             predict the development of sustained attention for negative
             emotion stimuli in soldiers returning from
             Iraq},
   Journal = {Clinical Psychological Science},
   Volume = {1},
   Number = {4},
   Pages = {413-425},
   Publisher = {SAGE Publications},
   Year = {2013},
   Month = {October},
   ISSN = {2167-7026},
   Abstract = {Biased attention toward negative stimuli is a known
             vulnerability for affective psychopathology. However,
             factors that contribute to the development of this cognitive
             bias are largely unknown. Variation within the serotonin
             transporter gene (i.e., 5-HTTLPR) is associated with
             increased susceptibility to environmental influence and
             biased processing of negative stimuli. Using a passive
             viewing eye-tracking paradigm, this study examined gaze
             fixation for emotion stimuli in 91 U.S. Army soldiers before
             and after deployment to Iraq. In addition, participants
             underwent genetic assay and provided in situ measures of war
             zone stress exposure. 5-HTTLPR short allele homozygotes were
             more likely than other genotype groups to develop a gaze
             bias toward negative stimuli as a function of increasing war
             zone stress, even when controlling for postdeployment
             posttraumatic stress disorder and depression severity. Short
             allele homozygotes appear especially sensitive to
             environmental influence, which likely contributes to the
             development of cognitive vulnerability to anxiety and mood
             disorders. © The Author(s) 2013.},
   Doi = {10.1177/2167702613485564},
   Key = {fds251968}
}

@article{Hyde2013,
   Author = {Hyde, LW and Shaw, DS and Hariri, AR},
   Title = {Understanding Youth Antisocial Behavior Using Neuroscience
             through a Developmental Psychopathology Lens: Review,
             Integration, and Directions for Research.},
   Journal = {Developmental review : DR},
   Volume = {33},
   Number = {3},
   Pages = {168-223},
   Publisher = {Elsevier BV},
   Address = {University of Michigan.},
   Year = {2013},
   Month = {September},
   ISSN = {0273-2297},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/24273368},
   Abstract = {Youth antisocial behavior (AB) is an important public health
             concern impacting perpetrators, victims, and society.
             Functional neuroimaging is becoming a more common and useful
             modality for understanding neural correlates of youth AB.
             Although there has been a recent increase in neuroimaging
             studies of youth AB and corresponding theoretical articles
             on the neurobiology of AB, there has been little work
             critically examining the strengths and weaknesses of
             individual studies and using this knowledge to inform the
             design of future studies. Additionally, research on
             neuroimaging and youth AB has not been integrated within the
             broader framework of developmental psychopathology. Thus,
             this paper provides an in-depth review of the youth AB
             functional neuroimaging literature with the following goals:
             1. to evaluate how this literature has informed our
             understanding of youth AB, 2. to evaluate current
             neuroimaging studies of youth AB from a developmental
             psychopathology perspective with a focus on integrating
             research from neuroscience and developmental
             psychopathology, as well as placing this research in the
             context of other related areas (e.g., psychopathy, molecular
             genetics), and 3. to examine strengths and weaknesses of
             neuroimaging and behavioral studies of youth AB to suggest
             how future studies can develop a more informed and
             integrated understanding of youth AB.},
   Language = {Eng},
   Doi = {10.1016/j.dr.2013.06.001},
   Key = {Hyde2013}
}

@article{Nikolova2013,
   Author = {Nikolova, YS and Singhi, EK and Drabant, EM and Hariri,
             AR},
   Title = {Reward-related ventral striatum reactivity mediates
             gender-specific effects of a galanin remote enhancer
             haplotype on problem drinking.},
   Journal = {Genes, brain, and behavior},
   Volume = {12},
   Number = {5},
   Pages = {516-524},
   Address = {Laboratory of NeuroGenetics, Department of Psychology and
             Neuroscience; Institute for Genome Sciences \& Policy, Duke
             University, Durham, NC 27705, USA. yuliya.nikolova@duke.edu},
   Year = {2013},
   Month = {July},
   ISSN = {1601-1848},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23489876},
   Abstract = {The neuropeptide galanin has been implicated in the
             regulation of appetitive and consummatory behaviors. Prior
             studies have shown that direct injection of galanin into the
             hypothalamus results in increased release of dopamine (DA)
             in the nucleus accumbens (NAcc), and parallel increases in
             food and alcohol consumption. These studies are consistent
             with a role of hypothalamic galanin in regulating reward
             system reactivity. In humans, a common functional haplotype
             (GAL5.1) within a remote enhancer region upstream of the
             galanin gene (GAL) affects promoter activity and galanin
             expression in hypothalamic neurons in vitro. Given the
             effects of hypothalamic galanin on NAcc DA release and the
             effects of the GAL5.1 haplotype on GAL expression, we
             examined the impact of this functional genetic variation on
             human reward-related ventral striatum (VS) reactivity. Using
             an imaging genetics strategy in Caucasian individuals
             (N = 138, 72 women) participating in the ongoing Duke
             Neurogenetics Study, we report a significant
             gender-by-genotype interaction (right hemisphere: F1,134
              = 8.08, P = 0.005; left hemisphere: F1,134
              = 5.39, P = 0.022), such that homozygosity for the
             GG haplotype, which predicts greater GAL expression, is
             associated with relatively increased VS reactivity in women
             (n = 50, right hemisphere: P = 0.015; left
             hemisphere: P = 0.060), but not in men (N = 49,
             P-values > 0.10). Furthermore, these differences in VS
             reactivity correlated positively with differences in alcohol
             use, such that VS reactivity mediated a gender-specific
             association between GAL5.1 haplotype and problem drinking.
             Our current results support those in animal models
             implicating galanin signaling in neural pathways associated
             with appetitive and consummatory behaviors of relevance for
             understanding risk for substance use and
             abuse.},
   Language = {eng},
   Doi = {10.1111/gbb.12035},
   Key = {Nikolova2013}
}

@article{Zuurbier2013,
   Author = {Zuurbier, LA and Nikolova, YS and Åhs, F and Hariri,
             AR},
   Title = {Uncinate fasciculus fractional anisotropy correlates with
             typical use of reappraisal in women but not
             men.},
   Journal = {Emotion (Washington, D.C.)},
   Volume = {13},
   Number = {3},
   Pages = {385-390},
   Address = {Laboratory of NeuroGenetics, Department of Psychology \&
             Neuroscience, Institute for Genome Sciences \& Policy, Duke
             University, Durham, NC, USA.},
   Year = {2013},
   Month = {June},
   ISSN = {1528-3542},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23398586},
   Abstract = {Emotion regulation refers to strategies through which
             individuals influence their experience and expression of
             emotions. Two typical strategies are reappraisal, a
             cognitive strategy for reframing the context of an emotional
             experience, and suppression, a behavioral strategy for
             inhibiting emotional responses. Functional neuroimaging
             studies have revealed that regions of the prefrontal cortex
             modulate amygdala reactivity during both strategies, but
             relatively greater downregulation of the amygdala occurs
             during reappraisal. Moreover, these studies demonstrated
             that engagement of this modulatory circuitry varies as a
             function of gender. The uncinate fasciculus is a major
             structural pathway connecting regions of the anterior
             temporal lobe, including the amygdala to inferior frontal
             regions, especially the orbitofrontal cortex. The objective
             of the current study was to map variability in the
             structural integrity of the uncinate fasciculus onto
             individual differences in self-reported typical use of
             reappraisal and suppression. Diffusion tensor imaging was
             used in 194 young adults to derive regional fractional
             anisotropy values for the right and left uncinate
             fasciculus. All participants also completed the Emotion
             Regulation Questionnaire. In women but not men,
             self-reported typical reappraisal use was positively
             correlated with fractional anisotropy values in a region of
             the left uncinate fasciculus within the orbitofrontal
             cortex. In contrast, typical use of suppression was not
             significantly correlated with fractional anisotropy in any
             region of the uncinate fasciculus in either men or women.
             Our data suggest that in women typical reappraisal use is
             specifically related to the integrity of white matter
             pathways linking the amygdala and prefrontal
             cortex.},
   Language = {eng},
   Doi = {10.1037/a0031163},
   Key = {Zuurbier2013}
}

@article{Kienast2013,
   Author = {Kienast, T and Schlagenhauf, F and Rapp, MA and Wrase, J and Daig, I and Buchholz, H-G and Smolka, MN and Gründer, G and Kumakura, Y and Cumming, P and Charlet, K and Bartenstein, P and Hariri, AR and Heinz,
             A},
   Title = {Dopamine-modulated aversive emotion processing fails in
             alcohol-dependent patients.},
   Journal = {Pharmacopsychiatry},
   Volume = {46},
   Number = {4},
   Pages = {130-136},
   Address = {Department of Psychiatry and Psychotherapy, Campus Charite
             Mitte, -Charite - Universitatsmedizin Berlin, Berlin,
             Germany.},
   Year = {2013},
   Month = {June},
   ISSN = {0176-3679},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23364872},
   Abstract = {Negative mood states after alcohol detoxification may
             enhance the relapse risk. As recently shown in healthy
             volunteers, dopamine storage capacity (V d) in the left
             amygdala was positively correlated with functional
             activation in the left amygdala and anterior cingulate
             cortex (ACC) during an emotional task; high functional
             connectivity between the amygdala and the ACC, a region
             important for emotion regulation, was associated with low
             trait anxiety. Based on these findings, we now tested
             whether detoxified alcohol-dependent patients have a
             disrupted modulation of the anterior cingulate cortex
             activation in response to aversive stimuli by amygdala
             dopamine. Furthermore, we asked whether disrupted functional
             coupling between amygdala and ACC during aversive processing
             is related to trait anxiety.We used combined
             6-[18F]-fluoro-l-DOPA positron emission tomography (PET),
             functional magnetic resonance imaging (fMRI) and
             Spielberger's state-trait anxiety questionnaire (STAI) in 11
             male detoxified alcohol-dependent patients compared to 13
             matched healthy controls.Unlike healthy controls, patients
             showed no significant correlation between our PET metric for
             dopamine storage capacity (FDOPA V d), in left amygdala and
             activation in left ACC. Moreover, the functional
             connectivity between amygdala and ACC during processing of
             aversive emotional stimuli was reduced in patients.
             Voxel-based morphometry did not reveal any discernible group
             differences in amygdala volume.These results suggest that
             dopamine-modulated corticolimbic circuit function is
             important for responding to emotional information such that
             apparent functional deficits in this neuromodulatory
             circuitry may contribute to trait anxiety in
             alcohol-dependent patients.},
   Language = {eng},
   Doi = {10.1055/s-0032-1331747},
   Key = {Kienast2013}
}

@article{Prather2013,
   Author = {Prather, AA and Bogdan, R and Hariri, AR},
   Title = {Impact of sleep quality on amygdala reactivity, negative
             affect, and perceived stress.},
   Journal = {Psychosomatic medicine},
   Volume = {75},
   Number = {4},
   Pages = {350-358},
   Address = {Department of Psychiatry, University of California, San
             Francisco, CA, USA. prathera@chc.ucsf.edu},
   Year = {2013},
   Month = {May},
   ISSN = {0033-3174},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23592753},
   Keywords = {Adolescent • Adult • Affect/*physiology •
             Amygdala/*physiopathology • Anger •
             Anxiety/physiopathology/psychology •
             Depression/physiopathology/psychology • Facial
             Expression • Fear • Female • Humans •
             *Magnetic Resonance Imaging • Male • Self Report
             • Severity of Illness Index • Sex Factors •
             Sleep/*physiology • Sleep Disorders/*physiopathology/psychology
             • Stress, Psychological/etiology/*physiopathology
             • Visual Cortex/physiopathology • Young
             Adult},
   Abstract = {<h4>Objective</h4>Research demonstrates a negative impact of
             sleep disturbance on mood and affect; however, the
             biological mechanisms mediating these links are poorly
             understood. Amygdala reactivity to negative stimuli has
             emerged as one potential pathway. Here, we investigate the
             influence of self-reported sleep quality on associations
             between threat-related amygdala reactivity and measures of
             negative affect and perceived stress.<h4>Methods</h4>Analyses
             on data from 299 participants (125 men, 50.5% white, mean
             [standard deviation] age = 19.6 [1.3] years) who completed
             the Duke Neurogenetics Study were conducted. Participants
             completed several self-report measures of negative affect
             and perceived stress. Threat-related (i.e., angry and
             fearful facial expressions) amygdala reactivity was assayed
             using blood oxygen level-dependent functional magnetic
             resonance imaging. Global sleep quality was assessed using
             the Pittsburgh Sleep Quality Index.<h4>Results</h4>Amygdala
             reactivity to fearful facial expressions predicted greater
             depressive symptoms and higher perceived stress in poor (β
             values = 0.18-1.86, p values < .05) but not good sleepers
             (β values = -0.13 to -0.01, p values > .05). In
             sex-specific analyses, men reporting poorer global sleep
             quality showed a significant association between amygdala
             reactivity and levels of depression and perceived stress (β
             values = 0.29-0.44, p values < .05). In contrast, no
             significant associations were observed in men reporting good
             global sleep quality or in women, irrespective of sleep
             quality.<h4>Conclusions</h4>This study provides novel
             evidence that self-reported sleep quality moderates the
             relationships between amygdala reactivity, negative affect,
             and perceived stress, particularly among
             men.},
   Language = {eng},
   Doi = {10.1097/psy.0b013e31828ef15b},
   Key = {Prather2013}
}

@article{Bogdan2013,
   Author = {Bogdan, R and Hyde, LW and Hariri, AR},
   Title = {A neurogenetics approach to understanding individual
             differences in brain, behavior, and risk for
             psychopathology.},
   Journal = {Molecular psychiatry},
   Volume = {18},
   Number = {3},
   Pages = {288-299},
   Address = {Laboratory of NeuroGenetics, Department of Psychology and
             Neuroscience, Duke University, Durham, NC 27705, USA.
             ryan.bogdan@duke.edu},
   Year = {2013},
   Month = {March},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22614291},
   Abstract = {Neurogenetics research has begun to advance our
             understanding of how genetic variation gives rise to
             individual differences in brain function, which, in turn,
             shapes behavior and risk for psychopathology. Despite these
             advancements, neurogenetics research is currently confronted
             by three major challenges: (1) conducting research on
             individual variables with small effects, (2) absence of
             detailed mechanisms, and (3) a need to translate findings
             toward greater clinical relevance. In this review, we
             showcase techniques and developments that address these
             challenges and highlight the benefits of a neurogenetics
             approach to understanding brain, behavior and
             psychopathology. To address the challenge of small effects,
             we explore approaches including incorporating the
             environment, modeling epistatic relationships and using
             multilocus profiles. To address the challenge of mechanism,
             we explore how non-human animal research, epigenetics
             research and genome-wide association studies can inform our
             mechanistic understanding of behaviorally relevant brain
             function. Finally, to address the challenge of clinical
             relevance, we examine how neurogenetics research can
             identify novel therapeutic targets and for whom treatments
             work best. By addressing these challenges, neurogenetics
             research is poised to exponentially increase our
             understanding of how genetic variation interacts with the
             environment to shape the brain, behavior and risk for
             psychopathology.},
   Language = {eng},
   Doi = {10.1038/mp.2012.35},
   Key = {Bogdan2013}
}

@article{fds251962,
   Author = {Salazar, E and Bogdan, R and Gorka, A and Hariri, AR and Carin,
             L},
   Title = {Exploring the mind: Integrating questionnaires and
             fMRI},
   Journal = {30th International Conference on Machine Learning, ICML
             2013},
   Number = {PART 2},
   Pages = {921-929},
   Year = {2013},
   Month = {January},
   Abstract = {A new model is developed for joint analysis of ordered,
             categorical, real and count data. The ordered and
             categorical data are answers to questionnaires, the (word)
             count data correspond to the text questions from the
             questionnaires, and the real data correspond to fMRI
             responses for each subject. The Bayesian model employs the
             von Mises distribution in a novel manner to infer sparse
             graphical models jointly across people, questions, fMRI
             stimuli and brain region, with this integrated within a new
             matrix factorization based on latent binary features. The
             model is compared with simpler alternatives on two real
             datasets. We also demonstrate the ability to predict the
             response of the brain to visual stimuli (as measured by
             fMRI), based on knowledge of how the associated person
             answered classical questionnaires. Copyright 2013 by the
             author(s).},
   Key = {fds251962}
}

@article{Fisher2013,
   Author = {Fisher, PM and Hariri, AR},
   Title = {Identifying serotonergic mechanisms underlying the
             corticolimbic response to threat in humans.},
   Journal = {Philosophical transactions of the Royal Society of London.
             Series B, Biological sciences},
   Volume = {368},
   Number = {1615},
   Pages = {20120192},
   Address = {Center for Integrated Molecular Brain Imaging, University of
             Copenhagen, Copenhagen 2100, Denmark. patrick.fisher@nru.dk},
   Year = {2013},
   Month = {January},
   ISSN = {0962-8436},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23440464},
   Keywords = {Amygdala/drug effects/*metabolism/physiology •
             Antidepressive Agents/pharmacology • Depression/drug
             therapy/physiopathology • Fear/*physiology •
             Humans • Neural Pathways/metabolism/physiology •
             Neuroimaging • Personality/physiology • Prefrontal
             Cortex/*metabolism/physiology • Protein Binding •
             Receptor, Serotonin, 5-HT1A/metabolism •
             Serotonin/*metabolism • Serotonin Uptake
             Inhibitors/pharmacology},
   Abstract = {A corticolimbic circuit including the amygdala and medial
             prefrontal cortex (mPFC) plays an important role in
             regulating sensitivity to threat, which is heightened in
             mood and anxiety disorders. Serotonin is a potent
             neuromodulator of this circuit; however, specific
             serotonergic mechanisms mediating these effects are not
             fully understood. Recent studies have evaluated molecular
             mechanisms mediating the effects of serotonin signalling on
             corticolimbic circuit function using a multi-modal
             neuroimaging strategy incorporating positron emission
             tomography and blood oxygen level-dependent functional
             magnetic resonance imaging. This multi-modal neuroimaging
             strategy can be integrated with additional techniques
             including imaging genetics and pharmacological challenge
             paradigms to more clearly understand how serotonin
             signalling modulates neural pathways underlying sensitivity
             to threat. Integrating these methodological approaches
             offers novel opportunities to identify mechanisms through
             which serotonin signalling contributes to differences in
             brain function and behaviour, which in turn can illuminate
             factors that confer risk for illness and inform the
             development of more effective treatment strategies.},
   Language = {eng},
   Doi = {10.1098/rstb.2012.0192},
   Key = {Fisher2013}
}

@article{Goetz2013,
   Author = {Goetz, E. L. and Hariri, A. R. and Pizzagalli, D. A. and Strauman, T. J.},
   Title = {Genetic moderation of the association between regulatory
             focus and reward responsiveness: a proof-of-concept
             study},
   Journal = {Biology of mood \& anxiety disorders},
   Volume = {3},
   Number = {1},
   Pages = {3},
   Address = {Department of Psychology \& Neuroscience, Duke University,
             Durham, NC, USA. elena.goetz@duke.edu.},
   Year = {2013},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23369671},
   Abstract = {BACKGROUND: Recent studies implicate individual differences
             in regulatory focus as contributing to self-regulatory
             dysfunction, particularly not responding to positive
             outcomes. How such individual differences emerge, however,
             is unclear. We conducted a proof-of-concept study to examine
             the moderating effects of genetically driven variation in
             dopamine signaling, a key modulator of neural reward
             circuits, on the association between regulatory focus and
             reward cue responsiveness. METHOD: Healthy Caucasians (N=59)
             completed a measure of chronic regulatory focus and a
             probabilistic reward task. A common functional genetic
             polymorphism impacting prefrontal dopamine signaling (COMT
             rs4680) was evaluated. RESULTS: Response bias, the
             participants' propensity to modulate behavior as a function
             of reward, was predicted by an interaction of regulatory
             focus and COMT genotype. Specifically, self-perceived
             success at achieving promotion goals predicted total
             response bias, but only for individuals with the COMT
             genotype (Val/Val) associated with relatively increased
             phasic dopamine signaling and cognitive flexibility.
             CONCLUSIONS: The combination of success in promotion goal
             pursuit and Val/Val genotype appears to facilitate
             responding to reward opportunities in the environment. This
             study is among the first to integrate an assessment of
             self-regulatory style with an examination of genetic
             variability that underlies responsiveness to positive
             outcomes in goal pursuit.},
   Language = {eng},
   Doi = {10.1186/2045-5380-3-3},
   Key = {Goetz2013}
}

@article{Carre2013a,
   Author = {Carre, J. M. and Murphy, K. R. and Hariri, A.
             R.},
   Title = {What lies beneath the face of aggression?},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {8},
   Number = {2},
   Pages = {224--9},
   Address = {Laboratory of NeuroGenetics, Department of Psychology and
             Neuroscience, Duke University, Durham, NC, USA.
             justin@carrelab.com},
   Year = {2013},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22198969},
   Keywords = {Adult • Aggression/*physiology •
             Amygdala/*physiology • Anger/physiology •
             Anthropometry • *Face/anatomy \& histology/physiology
             • *Facial Expression • Fear/physiology •
             Female • Humans • Magnetic Resonance
             Imaging/instrumentation/*methods • Male •
             Neuroimaging/methods/psychology • Neuropsychological
             Tests • Sex Characteristics • Young
             Adult},
   Abstract = {Recent evidence indicates that a sexually dimorphic feature
             of humans, the facial width-to-height ratio (FWHR), is
             positively correlated with reactive aggression, particularly
             in men. Also, predictions about the aggressive tendencies of
             others faithfully map onto FWHR in the absence of explicit
             awareness of this metric. Here, we provide the first
             evidence that amygdala reactivity to social signals of
             interpersonal challenge may underlie the link between
             aggression and the FWHR. Specifically, amygdala reactivity
             to angry faces was positively correlated with aggression,
             but only among men with relatively large FWHRs. The patterns
             of association were specific to angry facial expressions and
             unique to men. These links may reflect the common influence
             of pubertal testosterone on craniofacial growth and
             development of neural circuitry underlying aggression.
             Amygdala reactivity may also represent a plausible pathway
             through which FWHR may have evolved to represent an honest
             indicator of conspecific threat, namely by reflecting the
             responsiveness of neural circuitry mediating aggressive
             behavior.},
   Language = {eng},
   Doi = {10.1093/scan/nsr096},
   Key = {Carre2013a}
}

@article{Gianaros2013,
   Author = {Gianaros, PJ and Marsland, AL and Kuan, DC-H and Schirda, BL and Jennings, JR and Sheu, LK and Hariri, AR and Gross, JJ and Manuck,
             SB},
   Title = {An Inflammatory Pathway Links Atherosclerotic Cardiovascular
             Disease Risk to Neural Activity Evoked by the Cognitive
             Regulation of Emotion},
   Journal = {Biological Psychiatry},
   Volume = {75},
   Number = {9},
   Pages = {738-745},
   Publisher = {Elsevier BV},
   Address = {Department of Psychology (PJG, ALM, DC-HK, LKS, SBM).
             Electronic address: gianaros@pitt.edu.},
   Year = {2013},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/24267410},
   Abstract = {BACKGROUND: Cognitive reappraisal is a form of emotion
             regulation that alters emotional responding by changing the
             meaning of emotional stimuli. Reappraisal engages regions of
             the prefrontal cortex that support multiple functions,
             including visceral control functions implicated in
             regulating the immune system. Immune activity plays a role
             in the preclinical pathophysiology of atherosclerotic
             cardiovascular disease (CVD), an inflammatory condition that
             is highly comorbid with affective disorders characterized by
             problems with emotion regulation. Here, we tested whether
             prefrontal engagement by reappraisal would be associated
             with atherosclerotic CVD risk and whether this association
             would be mediated by inflammatory activity. METHODS:
             Community volunteers (n = 157; 30-54 years of age; 80 women)
             without DSM-IV Axis-1 psychiatric diagnoses or
             cardiovascular or immune disorders performed a functional
             neuroimaging task involving the reappraisal of negative
             emotional stimuli. Carotid artery intima-media thickness and
             inter-adventitial diameter were measured by ultrasonography
             and used as markers of preclinical atherosclerosis. Also
             measured were circulating levels of interleukin-6 (IL-6), an
             inflammatory cytokine linked to CVD risk and prefrontal
             neural activity. RESULTS: Greater reappraisal-related
             engagement of the dorsal anterior cingulate cortex was
             associated with greater preclinical atherosclerosis and
             IL-6. Moreover, IL-6 mediated the association of dorsal
             anterior cingulate cortex engagement with preclinical
             atherosclerosis. These results were independent of age, sex,
             race, smoking status, and other known CVD risk factors.
             CONCLUSIONS: The cognitive regulation of emotion might
             relate to CVD risk through a pathway involving the
             functional interplay between the anterior cingulate region
             of the prefrontal cortex and inflammatory
             activity.},
   Language = {Eng},
   Doi = {10.1016/j.biopsych.2013.10.012},
   Key = {Gianaros2013}
}

@article{Bogdan2012b,
   Author = {Bogdan, R and Hariri, AR},
   Title = {Neural embedding of stress reactivity.},
   Journal = {Nature neuroscience},
   Volume = {15},
   Number = {12},
   Pages = {1605-1607},
   Year = {2012},
   Month = {December},
   ISSN = {1097-6256},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23187689},
   Keywords = {Adolescent Behavior/*physiology • Amygdala/*growth \&
             development • Anxiety/*metabolism •
             Depression/*metabolism • Female • Humans •
             Hydrocortisone/*metabolism • Male • Prefrontal
             Cortex/*growth \& development},
   Language = {eng},
   Doi = {10.1038/nn.3270},
   Key = {Bogdan2012b}
}

@article{White2012,
   Author = {White, MG and Bogdan, R and Fisher, PM and Muñoz, KE and Williamson,
             DE and Hariri, AR},
   Title = {FKBP5 and emotional neglect interact to predict individual
             differences in amygdala reactivity.},
   Journal = {Genes, brain, and behavior},
   Volume = {11},
   Number = {7},
   Pages = {869-878},
   Address = {Laboratory of NeuroGenetics, Department of Psychology \&
             Neuroscience, Duke University, Durham, NC,
             USA.},
   Year = {2012},
   Month = {October},
   ISSN = {1601-1848},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22979952},
   Keywords = {Adolescent • Alleles • Amygdala/pathology/*physiopathology
             • *Child Abuse • Female • Gene-Environment
             Interaction • Haplotypes • Humans • Magnetic
             Resonance Imaging • Polymorphism, Single Nucleotide
             • Risk Factors • Stress, Psychological/pathology
             • Tacrolimus Binding Proteins/*genetics},
   Abstract = {Individual variation in physiological responsiveness to
             stress mediates risk for mental illness and is influenced by
             both experiential and genetic factors. Common polymorphisms
             in the human gene for FK506 binding protein 5 (FKBP5), which
             is involved in transcriptional regulation of the
             hypothalamic-pituitary-adrenal (HPA) axis, have been shown
             to interact with childhood abuse and trauma to predict
             stress-related psychopathology. In the current study, we
             examined if such gene-environment interaction effects may be
             related to variability in the threat-related reactivity of
             the amygdala, which plays a critical role in mediating
             physiological and behavioral adaptations to stress including
             modulation of the HPA axis. To this end, 139 healthy
             Caucasian youth completed a blood oxygen level-dependent
             functional magnetic resonance imaging probe of amygdala
             reactivity and self-report assessments of emotional neglect
             (EN) and other forms of maltreatment. These individuals were
             genotyped for 6 FKBP5 polymorphisms (rs7748266, rs1360780,
             rs9296158, rs3800373, rs9470080 and rs9394309) previously
             associated with psychopathology and/or HPA axis function.
             Interactions between each SNP and EN emerged such that risk
             alleles predicted relatively increased dorsal amygdala
             reactivity in the context of higher EN, even after
             correcting for multiple testing. Two different haplotype
             analyses confirmed this relationship as haplotypes with risk
             alleles also exhibited increased amygdala reactivity in the
             context of higher EN. Our results suggest that increased
             threat-related amygdala reactivity may represent a mechanism
             linking psychopathology to interactions between common
             genetic variants affecting HPA axis function and childhood
             trauma.},
   Language = {eng},
   Doi = {10.1111/j.1601-183x.2012.00837.x},
   Key = {White2012}
}

@article{Fisher2012,
   Author = {Fisher, PM and Hariri, AR},
   Title = {Linking variability in brain chemistry and circuit function
             through multimodal human neuroimaging.},
   Journal = {Genes, brain, and behavior},
   Volume = {11},
   Number = {6},
   Pages = {633-642},
   Address = {Center for Integrated Molecular Brain Imaging, Copenhagen
             University Hospital Rigshospitalet, Copenhagen, Denmark.
             patrick.fisher@gmail.com},
   Year = {2012},
   Month = {August},
   ISSN = {1601-1848},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22443230},
   Keywords = {Brain/physiology/radionuclide imaging • Brain
             Chemistry/genetics/*physiology • Brain
             Diseases/genetics/*physiopathology/*radionuclide imaging
             • Humans • Magnetic Resonance Imaging/methods
             • Neuroimaging/*methods • Positron-Emission
             Tomography/*methods},
   Abstract = {Identifying neurobiological mechanisms mediating the
             emergence of individual differences in behavior is critical
             for advancing our understanding of relative risk for
             psychopathology. Neuroreceptor positron emission tomography
             (PET) and functional magnetic resonance imaging (fMRI) can
             be used to assay in vivo regional brain chemistry and
             function, respectively. Typically, these neuroimaging
             modalities are implemented independently despite the
             capacity for integrated data sets to offer unique insight
             into molecular mechanisms associated with brain function.
             Through examples from the serotonin and dopamine system and
             its effects on threat- and reward-related brain function, we
             review evidence for how such a multimodal neuroimaging
             strategy can be successfully implemented. Furthermore, we
             discuss how multimodal PET-fMRI can be integrated with
             techniques such as imaging genetics, pharmacological
             challenge paradigms and gene-environment interaction models
             to more completely map biological pathways mediating
             individual differences in behavior and related risk for
             psychopathology and inform the development of novel
             therapeutic targets.},
   Language = {eng},
   Doi = {10.1111/j.1601-183x.2012.00786.x},
   Key = {Fisher2012}
}

@article{Hariri2012,
   Author = {Hariri, AR},
   Title = {The highs and lows of amygdala reactivity in bipolar
             disorders.},
   Journal = {The American journal of psychiatry},
   Volume = {169},
   Number = {8},
   Pages = {780-783},
   Year = {2012},
   Month = {August},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22854927},
   Keywords = {Bipolar Disorder/*physiopathology •
             Brain/*physiopathology • Emotions/*physiology •
             Female • Humans • *Magnetic Resonance Imaging
             • Male},
   Language = {eng},
   Doi = {10.1176/appi.ajp.2012.12050639},
   Key = {Hariri2012}
}

@article{Brown2012,
   Author = {Brown, AA and Jensen, J and Nikolova, YS and Djurovic, S and Agartz, I and Server, A and Ferrell, RE and Manuck, SB and Mattingsdal, M and Melle,
             I and Hariri, AR and Frigessi, A and Andreassen, OA},
   Title = {Genetic variants affecting the neural processing of human
             facial expressions: evidence using a genome-wide functional
             imaging approach.},
   Journal = {Translational psychiatry},
   Volume = {2},
   Pages = {e143},
   Address = {Division of Mental Health and Addiction, Institute of
             Clinical Medicine, University of Oslo, Oslo, Norway.
             andrew.brown@medisin.uio.no},
   Year = {2012},
   Month = {July},
   ISSN = {2158-3188},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22828495},
   Keywords = {Adult • Brain Mapping • *Facial Expression •
             Female • Genetic Variation • Genome-Wide
             Association Study • Humans • Magnetic Resonance
             Imaging/methods • Male • Polymorphism, Single
             Nucleotide • Reproducibility of Results • Temporal
             Lobe/*physiopathology • Visual Perception/*genetics},
   Abstract = {Human faces present crucial visual information for social
             interaction. Specialized brain regions are involved in the
             perception of faces, with the fusiform face area (FFA) a key
             neuronal substrate. Face processing is genetically
             controlled, but by which specific genes is unknown. A
             genome-wide approach identified common single nucleotide
             polymorphisms (SNPs) associated with areas of increased
             brain activity in response to affective facial expressions,
             measured with functional magnetic resonance imaging. SNPs in
             20 genetic regions were linked with neural responses to
             negative facial expressions in a Norwegian sample (n=246),
             which included patients with mental illness. Three genetic
             regions were linked with FFA activation in a further
             discovery experiment using positive facial expressions and
             involving many of the same individuals (n=284). Two of these
             three regions showed significant association with right FFA
             activation to negative facial expressions in an independent
             North American replication sample of healthy Caucasians
             (n=85, 3q26.31, P=0.004; 20p12.3, P=0.045). The activation
             patterns were particularly striking for the SNP in 3q26.31,
             which lies in a gene TMEM212; only the FFA was activated.
             The specialized function of this brain region suggests that
             TMEM212 could contribute to the innate architecture of face
             processing.},
   Language = {eng},
   Doi = {10.1038/tp.2012.67},
   Key = {Brown2012}
}

@article{Nikolova2012b,
   Author = {Nikolova, YS and Bogdan, R and Brigidi, BD and Hariri,
             AR},
   Title = {Ventral striatum reactivity to reward and recent life stress
             interact to predict positive affect.},
   Journal = {Biological psychiatry},
   Volume = {72},
   Number = {2},
   Pages = {157-163},
   Address = {Department of Psychology and Neuroscience and Institute for
             Genome Sciences and Policy, Duke University, Durham, NC
             27705, USA. yuliya.nikolova@duke.edu},
   Year = {2012},
   Month = {July},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22534456},
   Keywords = {Adult • Affect/*physiology • Brain
             Mapping/methods/*psychology • Corpus Striatum/blood
             supply/*physiology • Depressive Disorder,
             Major/*physiopathology • Ethnic Groups/psychology
             • Female • Humans • Life Change Events •
             Magnetic Resonance Imaging/methods/psychology • Male
             • *Reward • Self Report • Stress,
             Psychological/*physiopathology},
   Abstract = {<h4>Background</h4>Stressful life events are among the most
             reliable precipitants of major depressive disorder; yet, not
             everyone exposed to stress develops depression. It has been
             hypothesized that robust neural reactivity to reward and
             associated stable levels of positive affect (PA) may protect
             against major depressive disorder in the context of
             environmental adversity. However, little empirical data
             exist to confirm this postulation. Here, we test the
             hypothesis that individuals with relatively low ventral
             striatum (VS) reactivity to reward will show low PA levels
             in the context of recent life stress, while those with
             relatively high VS reactivity will be protected against
             these potentially depressogenic effects.<h4>Methods</h4>Differential
             VS reactivity to positive feedback was assessed using blood
             oxygen level-dependent functional magnetic resonance imaging
             in a sample of 200 nonpatient young adults. Recent life
             stress, current depressive symptoms, and PA were assessed
             via self-report. Linear regression models were used to
             investigate the moderating effects of VS reactivity on the
             relationship between recent stress and state PA across
             participants.<h4>Results</h4>Recent life stress interacted
             with VS reactivity to predict self-reported state PA, such
             that higher levels of life stress were associated with lower
             PA for participants with relatively low, but not for those
             with high, VS reactivity. These effects were independent of
             age, gender, race/ethnicity, trait PA, and early childhood
             trauma.<h4>Conclusions</h4>The current results provide
             empirical evidence for the potentially protective role of
             robust reward-related neural responsiveness against
             reductions in PA that may occur in the wake of life stress
             and possibly vulnerability to depression precipitated by
             stressful life events.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2012.03.014},
   Key = {Nikolova2012b}
}

@article{Sweitzer2012,
   Author = {Sweitzer, MM and Donny, EC and Hariri, AR},
   Title = {Imaging genetics and the neurobiological basis of individual
             differences in vulnerability to addiction.},
   Journal = {Drug Alcohol Depend},
   Volume = {123 Suppl 1},
   Number = {SUPPL.1},
   Pages = {S59-S71},
   Address = {Department of Psychology, University of Pittsburgh,
             Pittsburgh, PA 15260, USA. mms74@pitt.edu},
   Year = {2012},
   Month = {June},
   ISSN = {0376-8716},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22342427},
   Keywords = {Adult • Basal Ganglia/drug effects • Behavior,
             Addictive/*genetics/physiopathology • Genetic
             Variation/*physiology • Humans • Impulsive
             Behavior/*genetics/psychology • Individuality •
             Male • Neural Pathways/*physiopathology •
             *Neuroimaging • Phenotype • Polymorphism,
             Genetic/physiology • Receptors, Dopamine
             D2/*genetics/physiology • Tobacco Use
             Disorder/*genetics/physiopathology},
   Abstract = {BACKGROUND: Addictive disorders are heritable, but the
             search for candidate functional polymorphisms playing an
             etiological role in addiction is hindered by complexity of
             the phenotype and the variety of factors interacting to
             impact behavior. Advances in human genome sequencing and
             neuroimaging technology provide an unprecedented opportunity
             to explore the impact of functional genetic variants on
             variability in behaviorally relevant neural circuitry. Here,
             we present a model for merging these technologies to trace
             the links between genes, brain, and addictive behavior.
             METHODS: We describe imaging genetics and discuss the
             utility of its application to addiction. We then review data
             pertaining to impulsivity and reward circuitry as an example
             of how genetic variation may lead to variation in behavioral
             phenotype. Finally, we present preliminary data relating the
             neural basis of reward processing to individual differences
             in nicotine dependence. RESULTS: Complex human behaviors
             such as addiction can be traced to their basic genetic
             building blocks by identifying intermediate behavioral
             phenotypes, associated neural circuitry, and underlying
             molecular signaling pathways. Impulsivity has been linked
             with variation in reward-related activation in the ventral
             striatum (VS), altered dopamine signaling, and functional
             polymorphisms of DRD2 and DAT1 genes. In smokers, changes in
             reward-related VS activation induced by smoking abstinence
             may be associated with severity of nicotine dependence.
             CONCLUSIONS: Variation in genes related to dopamine
             signaling may contribute to heterogeneity in VS sensitivity
             to reward and, ultimately, to addiction. These findings
             illustrate the utility of the imaging genetics approach for
             investigating the neurobiological basis for vulnerability to
             addiction.},
   Language = {eng},
   Doi = {10.1016/j.drugalcdep.2012.01.017},
   Key = {Sweitzer2012}
}

@article{Ousdal2012,
   Author = {Ousdal, OT and Anand Brown and A and Jensen, J and Nakstad, PH and Melle,
             I and Agartz, I and Djurovic, S and Bogdan, R and Hariri, AR and Andreassen, OA},
   Title = {Associations between variants near a monoaminergic pathways
             gene (PHOX2B) and amygdala reactivity: a genome-wide
             functional imaging study.},
   Journal = {Twin research and human genetics : the official journal of
             the International Society for Twin Studies},
   Volume = {15},
   Number = {3},
   Pages = {273-285},
   Address = {Oslo University Hospital, Oslo, Norway. o.t.ousdal@medisin.uio.no},
   Year = {2012},
   Month = {June},
   ISSN = {1832-4274},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22856363},
   Keywords = {Adult • Amygdala/*physiology • Bipolar
             Disorder/genetics/physiopathology •
             Catecholamines/biosynthesis • Female •
             *Genome-Wide Association Study • Genotype •
             Homeodomain Proteins/*genetics/metabolism • Humans
             • Linkage Disequilibrium • *Magnetic Resonance
             Imaging • Male • *Polymorphism, Single Nucleotide
             • Schizophrenia/genetics/physiopathology •
             Serotonin/biosynthesis • Signal Transduction/*genetics
             • Transcription Factors/*genetics/metabolism},
   Abstract = {As the amygdala is part of the phylogenetic old brain, and
             its anatomical and functional properties are conserved
             across species, it is reasonable to assume genetic influence
             on its activity. A large corpus of candidate gene studies
             indicate that individual differences in amygdala activity
             may be caused by genetic variants within monoaminergic
             signaling pathways such as dopamine, serotonin, and
             norepinephrine. However, to our knowledge, the use of
             genome-wide data to discover genetic variants underlying
             individual differences in adult amygdala activity is novel.
             In the present study, the combination of genome-wide data
             and functional imaging phenotypes from an emotional faces
             task yielded a significant association between rs10014254
             and the amygdala using a region of interest approach. This
             single nucleotide polymorphism is located in a regulatory
             region upstream of the Paired-like homeobox 2b (PHOX2B)
             gene; therefore it could affect the expression of this gene.
             PHOX2B regulates the expression of enzymes necessary for the
             synthesis of several monoamines and is essential for the
             development of the autonomic nervous system. However, an
             attempt to replicate the finding in an independent sample
             from North America did not succeed. The synthesis of
             functional magnetic resonance imaging (fMRI) and genome-wide
             data takes a hypothesis-free approach as to which genetic
             variants are of interest. Therefore, we believe that an
             undirected finding within such a plausible region is of
             interest, and that our results add further support to the
             hypothesis that monoaminergic signaling pathways play a
             central role in regulating amygdala activity.},
   Language = {eng},
   Doi = {10.1017/thg.2012.5},
   Key = {Ousdal2012}
}

@article{Josephs2012,
   Author = {Josephs, RA and Telch, MJ and Hixon, JG and Evans, JJ and Lee, H and Knopik, VS and McGeary, JE and Hariri, AR and Beevers,
             CG},
   Title = {Genetic and hormonal sensitivity to threat: testing a
             serotonin transporter genotype × testosterone
             interaction.},
   Journal = {Psychoneuroendocrinology},
   Volume = {37},
   Number = {6},
   Pages = {752-761},
   Address = {Department of Psychology, The University of Texas at Austin,
             Austin, TX 78756, USA. josephs@mail.psy.utexas.edu},
   Year = {2012},
   Month = {June},
   ISSN = {0306-4530},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21978869},
   Keywords = {Carbon Dioxide/diagnostic use • DNA/genetics •
             Depression/genetics/psychology • Ethnic Groups •
             Female • Genotype • Heterozygote • Humans
             • Hydrocortisone/blood • Male • Military
             Personnel • Psychiatric Status Rating Scales •
             Psychomotor Performance/physiology • Questionnaires
             • Saliva/chemistry • Serotonin Plasma Membrane
             Transport Proteins/*genetics/*metabolism • Social
             Environment • Stress, Psychological/*genetics/*metabolism
             • Testosterone/*blood • Young Adult},
   Abstract = {<h4>Background</h4>Striking parallels are observed when
             comparing the literature on the 5-HTTLPR of the serotonin
             transporter gene (SLC6A4) to the testosterone (T) literature
             on measures of stress reactivity and neural activity. Short
             (S) allele carriers and individuals higher in testosterone
             levels show exaggerated stress responses, amygdala
             hyperactivity, and reduction of amygdala-prefrontal cortex
             coupling when exposed to threat.<h4>Methods</h4>Three
             studies tested the hypothesis that higher T, S carriers
             would show increased cortisol responses to
             threat.<h4>Results</h4>Supporting the hypothesis, a T ×
             5-HTTLPR interaction was obtained across all studies.
             Threats to status via social exclusion (Study 1),
             cognitive/perceptual failure (Study 2), and physical
             competence (Study 3) all produced elevated cortisol levels
             in S carriers with higher T levels. An unexpected result was
             that 5-HTTLPR long (L) allele homozygotes with higher T
             showed lower cortisol levels in response to threat-a pattern
             of response that closely parallels that reported for
             psychopathic individuals. Finally, combining effect sizes
             across studies showed that the likelihood that these effects
             were due to Type 1 errors was quite low.<h4>Conclusions</h4>What
             emerges from these studies is a novel yet reliable, and
             synergistic relationship between 5-HTTLPR genotype and
             testosterone on stress reactivity, possibly conferring
             vulnerability for multiple neuropsychiatric
             disorders.},
   Language = {eng},
   Doi = {10.1016/j.psyneuen.2011.09.006},
   Key = {Josephs2012}
}

@article{Bogdan2012a,
   Author = {Bogdan, R and Williamson, DE and Hariri, AR},
   Title = {Mineralocorticoid receptor Iso/Val (rs5522) genotype
             moderates the association between previous childhood
             emotional neglect and amygdala reactivity.},
   Journal = {The American journal of psychiatry},
   Volume = {169},
   Number = {5},
   Pages = {515-522},
   Address = {Laboratory of NeuroGenetics, Department of Psychology and
             Neuroscience, and Institute for Genome Sciences and Policy,
             Duke University, Durham, NC, USA. bogdan.ryan@gmail.com},
   Year = {2012},
   Month = {May},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22407082},
   Keywords = {Adolescent • Alleles • Amygdala/*physiopathology
             • Child • *Child Abuse/psychology • Female
             • Functional Neuroimaging • Genotype •
             Homozygote • Humans • Magnetic Resonance Imaging
             • Male • Mutation, Missense/*genetics/physiology
             • Receptors, Mineralocorticoid/*genetics/physiology},
   Abstract = {<h4>Objective</h4>The amygdala is especially reactive to
             threatening stimuli, and the degree of reactivity predicts
             individual differences in the expression of depression and
             anxiety. Emerging research suggests that emotional neglect
             during childhood as well as hypercortisolemia may lead to
             heightened threat-related amygdala reactivity. This raises
             the possibility that genetic variation affecting
             hypothalamic-pituitary-adrenal (HPA) axis function
             contributes to individual differences in amygdala
             reactivity, both independently and as a function of
             childhood emotional neglect.<h4>Method</h4>This study
             assessed whether the mineralocorticoid receptor iso/val
             polymorphism (rs5522), a functional genetic variant
             affecting HPA axis function, influenced threat-related
             amygdala reactivity in 279 individuals in late childhood and
             early adolescence. The study also explored the extent to
             which any effects of the genotype on amygdala reactivity
             were contingent upon previous childhood emotional
             neglect.<h4>Results</h4>Prior childhood emotional neglect
             and the val allele were associated with greater amygdala
             reactivity. Moreover, a significant genotype-by-emotional
             neglect interaction was observed whereby greater amygdala
             reactivity in val allele carriers was independent of
             previous childhood emotional neglect, while greater
             reactivity in iso homozygotes was revealed only in the
             context of a history of elevated emotional neglect. At
             relatively low levels of previous emotional neglect, val
             carriers had heightened amygdala reactivity relative to iso
             homozygotes.<h4>Conclusions</h4>These results suggest that
             relatively greater amygdala reactivity may represent a
             biological mechanism through which childhood adversity and
             functional genetic variation in HPA axis responsiveness to
             stress may mediate risk for psychopathology.},
   Language = {eng},
   Doi = {10.1176/appi.ajp.2011.11060855},
   Key = {Bogdan2012a}
}

@article{Lahey2012a,
   Author = {Lahey, BB and McNealy, K and Knodt, A and Zald, DH and Sporns, O and Manuck, SB and Flory, JD and Applegate, B and Rathouz, PJ and Hariri,
             AR},
   Title = {Using confirmatory factor analysis to measure
             contemporaneous activation of defined neuronal networks in
             functional magnetic resonance imaging.},
   Journal = {NeuroImage},
   Volume = {60},
   Number = {4},
   Pages = {1982-1991},
   Address = {Department of Health Studies, University of Chicago,
             Chicago, IL 60637, United States. blahey@health.bsd.uchicago.edu},
   Year = {2012},
   Month = {May},
   ISSN = {1053-8119},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22348884},
   Keywords = {Adolescent • Adult • Brain/physiology • Brain
             Mapping/*methods • *Factor Analysis, Statistical •
             Female • Humans • Image Interpretation,
             Computer-Assisted/*methods • *Magnetic Resonance
             Imaging • Male • *Models, Theoretical • Young
             Adult},
   Abstract = {Functional neuroimaging often generates large amounts of
             data on regions of interest. Such data can be addressed
             effectively with a widely-used statistical technique based
             on measurement theory that has not yet been applied to
             neuroimaging. Confirmatory factor analysis is a convenient
             hypothesis-driven modeling environment that can be used to
             conduct formal statistical tests comparing alternative
             hypotheses regarding the elements of putative neuronal
             networks. In such models, measures of each activated region
             of interest are treated as indicators of an underlying
             latent construct that represents the contemporaneous
             activation of the elements in the network. As such,
             confirmatory factor analysis focuses analyses on the
             activation of hypothesized networks as a whole, improves
             statistical power by modeling measurement error, and
             provides a theory-based approach to data reduction with a
             robust statistical basis. This approach is illustrated using
             data on seven regions of interest in a hypothesized
             mesocorticostriatal reward system in a sample of 262 adult
             volunteers assessed during a card-guessing reward task. A
             latent construct reflecting contemporaneous activation of
             the reward system was found to be significantly associated
             with a latent construct measuring impulsivity, particularly
             in males.},
   Language = {eng},
   Doi = {10.1016/j.neuroimage.2012.02.002},
   Key = {Lahey2012a}
}

@article{Terburg2012,
   Author = {Terburg, D and Morgan, BE and Montoya, ER and Hooge, IT and Thornton,
             HB and Hariri, AR and Panksepp, J and Stein, DJ and van Honk,
             J},
   Title = {Hypervigilance for fear after basolateral amygdala damage in
             humans.},
   Journal = {Translational psychiatry},
   Volume = {2},
   Pages = {e115},
   Address = {Department of Psychology, Utrecht University, Utrecht, The
             Netherlands. d.terburg@uu.nl},
   Year = {2012},
   Month = {May},
   ISSN = {2158-3188},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22832959},
   Keywords = {Adult • Amygdala/*physiopathology •
             Anxiety/genetics/pathology/*physiopathology • Attention
             • Brain Damage, Chronic/genetics/pathology/*physiopathology
             • Brain Mapping • Calcinosis/genetics/pathology/physiopathology
             • Discrimination (Psychology)/physiology •
             Dominance, Cerebral/physiology • Emotions/physiology
             • Facial Expression • Fear/*physiology •
             Female • Humans • Image Interpretation,
             Computer-Assisted • Lipoid Proteinosis of Urbach and
             Wiethe/genetics/pathology/*physiopathology • Magnetic
             Resonance Imaging • Male • Middle Aged •
             Nerve Growth Factors • Neural Inhibition/*physiology
             • Pattern Recognition, Visual/physiology •
             Recognition (Psychology)/physiology • Reference Values
             • Stroop Test • Subliminal Stimulation},
   Abstract = {Recent rodent research has shown that the basolateral
             amygdala (BLA) inhibits unconditioned, or innate, fear. It
             is, however, unknown whether the BLA acts in similar ways in
             humans. In a group of five subjects with a rare genetic
             syndrome, that is, Urbach-Wiethe disease (UWD), we used a
             combination of structural and functional neuroimaging, and
             established focal, bilateral BLA damage, while other
             amygdala sub-regions are functionally intact. We tested the
             translational hypothesis that these BLA-damaged UWD-subjects
             are hypervigilant to facial expressions of fear, which are
             prototypical innate threat cues in humans. Our data indeed
             repeatedly confirm fear hypervigilance in these UWD
             subjects. They show hypervigilant responses to unconsciously
             presented fearful faces in a modified Stroop task. They
             attend longer to the eyes of dynamically displayed fearful
             faces in an eye-tracked emotion recognition task, and in
             that task recognize facial fear significantly better than
             control subjects. These findings provide the first direct
             evidence in humans in support of an inhibitory function of
             the BLA on the brain's threat vigilance system, which has
             important implications for the understanding of the
             amygdala's role in the disorders of fear and
             anxiety.},
   Language = {eng},
   Doi = {10.1038/tp.2012.46},
   Key = {Terburg2012}
}

@article{Joeyen-Waldorf2012,
   Author = {Joeyen-Waldorf, J and Nikolova, YS and Edgar, N and Walsh, C and Kota,
             R and Lewis, DA and Ferrell, R and Manuck, SB and Hariri, AR and Sibille,
             E},
   Title = {Adenylate cyclase 7 is implicated in the biology of
             depression and modulation of affective neural
             circuitry.},
   Journal = {Biological psychiatry},
   Volume = {71},
   Number = {7},
   Pages = {627-632},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pennsylvania 15219, USA.},
   Year = {2012},
   Month = {April},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22264442},
   Keywords = {Adenylate Cyclase/genetics/metabolism/*physiology •
             Adult • Amygdala/metabolism/*physiopathology •
             Animals • Depressive Disorder/genetics/metabolism/*physiopathology
             • Disease Models, Animal • Emotions/*physiology
             • Female • Functional Neuroimaging/methods/*psychology
             • Gene Expression/genetics/physiology • Genetic
             Predisposition to Disease • Genotype • Gyrus
             Cinguli/physiopathology • Humans • Magnetic
             Resonance Imaging/methods/psychology • Male • Mice
             • Mice, Inbred C57BL • Mice, Knockout •
             Polymorphism, Single Nucleotide • Serotonin Plasma
             Membrane Transport Proteins/genetics},
   Abstract = {<h4>Background</h4>Evolutionarily conserved genes and their
             associated molecular pathways can serve as a translational
             bridge between human and mouse research, extending our
             understanding of biological pathways mediating individual
             differences in behavior and risk for psychopathology.<h4>Methods</h4>Comparative
             gene array analysis in the amygdala and cingulate cortex
             between the serotonin transporter knockout mouse, a genetic
             animal model replicating features of human depression, and
             existing brain transcriptome data from postmortem tissue
             derived from clinically depressed humans was conducted to
             identify genes with similar changes across species (i.e.,
             conserved) that may help explain risk of depressive-like
             phenotypes. Human neuroimaging analysis was then used to
             investigate the impact of a common single-nucleotide
             polymorphism (rs1064448) in a gene with identified conserved
             human-mouse changes, adenylate cyclase 7 (ADCY7), on
             threat-associated amygdala reactivity in two large
             independent samples.<h4>Results</h4>Comparative analysis
             identified genes with conserved transcript changes in
             amygdala (n = 29) and cingulate cortex (n = 19), both
             critically involved in the generation and regulation of
             emotion. Selected results were confirmed by real-time
             quantitative polymerase chain reaction, including
             upregulation in the amygdala of transcripts for ADCY7, a
             gene previously implicated in human depression and
             associated with altered emotional responsiveness in mouse
             models. Translating these results back to living healthy
             human subjects, we show that genetic variation (rs1064448)
             in ADCY7 biases threat-related amygdala reactivity.<h4>Conclusions</h4>This
             converging cross-species evidence implicates ADCY7 in the
             modulation of mood regulatory neural mechanisms and,
             possibly, risk for and pathophysiology of depression,
             together supporting a continuous dimensional approach to
             major depressive disorder and other affective
             disorders.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2011.11.029},
   Key = {Joeyen-Waldorf2012}
}

@article{Drabant2012,
   Author = {Drabant, EM and Ramel, W and Edge, MD and Hyde, LW and Kuo, JR and Goldin,
             PR and Hariri, AR and Gross, JJ},
   Title = {Neural mechanisms underlying 5-HTTLPR-related sensitivity to
             acute stress.},
   Journal = {The American journal of psychiatry},
   Volume = {169},
   Number = {4},
   Pages = {397-405},
   Address = {Department of Psychology, Stanford University, Stanford,
             California, USA.},
   Year = {2012},
   Month = {April},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22362395},
   Keywords = {Adult • Alleles • Brain/*physiopathology •
             Female • Functional Neuroimaging/methods/*psychology
             • Genotype • Humans • Magnetic Resonance
             Imaging/methods/*psychology • Neural
             Pathways/physiopathology • Polymorphism, Single
             Nucleotide • Serotonin Plasma Membrane Transport
             Proteins/genetics/*physiology • Stress,
             Psychological/genetics/*physiopathology},
   Abstract = {<h4>Objective</h4>Many studies have shown that 5-HTTLPR
             genotype interacts with exposure to stress in conferring
             risk for psychopathology. However, the specific neural
             mechanisms through which this gene-by-environment
             interaction confers risk remain largely unknown, and no
             study to date has directly examined the modulatory effects
             of 5-HTTLPR on corticolimbic circuit responses during
             exposure to acute stress.<h4>Method</h4>An acute laboratory
             stressor was administered to 51 healthy women during
             blood-oxygen-level-dependent functional magnetic resonance
             imaging. In this task, participants were threatened with
             electric shocks of uncertain intensity, which were
             unpredictably delivered to the wrist after a long
             anticipatory cue period of unpredictable
             duration.<h4>Results</h4>Relative to women carrying the L
             allele, those with the SS genotype showed enhanced
             activation during threat anticipation in a network of
             regions, including the amygdala, hippocampus, anterior
             insula, thalamus, pulvinar, caudate, precuneus, anterior
             cingulate cortex, and medial prefrontal cortex. Individuals
             with the SS genotype also displayed enhanced positive
             coupling between medial prefrontal cortex activation and
             anxiety experience, whereas enhanced negative coupling
             between insula activation and perceived success at
             regulating anxiety was observed in individuals carrying the
             L allele.<h4>Conclusions</h4>These findings suggest that
             during stress exposure, neural systems that enhance fear and
             arousal, modulate attention toward threat, and perseverate
             on emotional salience of the threat may be engaged
             preferentially in individuals with the SS genotype. This may
             be one mechanism underlying the risk for psychopathology
             conferred by the S allele upon exposure to life
             stressors.},
   Language = {eng},
   Doi = {10.1176/appi.ajp.2011.10111699},
   Key = {Drabant2012}
}

@article{Carre2012,
   Author = {Carré, JM and Fisher, PM and Manuck, SB and Hariri,
             AR},
   Title = {Interaction between trait anxiety and trait anger predict
             amygdala reactivity to angry facial expressions in men but
             not women.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {7},
   Number = {2},
   Pages = {213-221},
   Address = {Department of Psychology and Neuroscience, Duke University,
             Durham, NC 27708, USA. justin.carre@duke.edu},
   Year = {2012},
   Month = {February},
   ISSN = {1749-5016},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21183456},
   Keywords = {Adult • Amygdala/*physiology • Anger/*physiology
             • Anxiety/*psychology • Brain/physiology •
             Emotions/physiology • *Facial Expression • Female
             • Humans • Individuality • Male • Middle
             Aged • Neuroimaging},
   Abstract = {The amygdala is critically involved in mediating
             physiological and behavioral responses to threat. In
             particular, neuroimaging research indicates that the
             amygdala is highly responsive to facial signals of threat
             such as fearful and angry expressions. However, individuals
             differ substantially in both their relative sensitivity to
             threat and the magnitude of amygdala reactivity to facial
             signals of threat. Here, we report the novel finding that
             individual differences in trait anger are positively
             correlated with bilateral dorsal amygdala reactivity to
             angry facial expressions, but only among men with elevated
             trait anxiety scores. These findings add to the growing body
             of evidence indicating that variability in personality
             traits contribute to individual differences in
             threat-related amygdala reactivity and further suggest that
             heightened amygdala reactivity to angry faces may be
             uniquely involved in the expression of reactive aggression
             in men.},
   Language = {eng},
   Doi = {10.1093/scan/nsq101},
   Key = {Carre2012}
}

@article{Davis2013,
   Author = {Davis, FC and Knodt, AR and Sporns, O and Lahey, BB and Zald, DH and Brigidi, BD and Hariri, AR},
   Title = {Impulsivity and the Modular Organization of Resting-State
             Neural Networks},
   Journal = {Cerebral cortex},
   Volume = {23},
   Number = {6},
   Pages = {1444-1452},
   Address = {Laboratory of NeuroGenetics, Department of Psychology and
             Neuroscience, Duke University, Durham, NC 27708, USA.
             caroline.davis@duke.edu},
   Year = {2012},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22645253},
   Keywords = {Adolescent • *Brain Mapping • Female • Head
             Movements • Humans • Image Processing,
             Computer-Assisted • Impulsive Behavior/classification/*pathology
             • Magnetic Resonance Imaging • Male • Models,
             Neurological • Neural Pathways/blood supply/*pathology
             • Oxygen • Rest/*physiology • Self Report
             • Young Adult},
   Abstract = {Impulsivity is a complex trait associated with a range of
             maladaptive behaviors, including many forms of
             psychopathology. Previous research has implicated multiple
             neural circuits and neurotransmitter systems in impulsive
             behavior, but the relationship between impulsivity and
             organization of whole-brain networks has not yet been
             explored. Using graph theory analyses, we characterized the
             relationship between impulsivity and the functional
             segregation (``modularity'') of the whole-brain network
             architecture derived from resting-state functional magnetic
             resonance imaging (fMRI) data. These analyses revealed
             remarkable differences in network organization across the
             impulsivity spectrum. Specifically, in highly impulsive
             individuals, regulatory structures including medial and
             lateral regions of the prefrontal cortex were isolated from
             subcortical structures associated with appetitive drive,
             whereas these brain areas clustered together within the same
             module in less impulsive individuals. Further exploration of
             the modular organization of whole-brain networks revealed
             novel shifts in the functional connectivity between visual,
             sensorimotor, cortical, and subcortical structures across
             the impulsivity spectrum. The current findings highlight the
             utility of graph theory analyses of resting-state fMRI data
             in furthering our understanding of the neurobiological
             architecture of complex behaviors.},
   Language = {eng},
   Doi = {10.1093/cercor/bhs126},
   Key = {Davis2013}
}

@article{Bogdan2012c,
   Author = {Bogdan, R and Carre, JM and Hariri, AR},
   Title = {Toward a mechanistic understanding of how variability in
             neurobiology shapes individual differences in
             behavior},
   Journal = {Current topics in behavioral neurosciences},
   Volume = {12},
   Pages = {361-393},
   Address = {Laboratory of NeuroGenetics, Department of Psychology \&
             Neuroscience, Institute for Genome Sciences \& Policy, Duke
             University, 417 Chapel Drive, Durham, NC, 27708, USA,
             bogdan.ryan@gmail.com.},
   Year = {2012},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22437943},
   Abstract = {Research has begun to identify how variability in brain
             function contributes to individual differences in complex
             behavioral traits. Examining variability in molecular
             signaling pathways with emerging and established
             methodologies such as pharmacologic fMRI, multimodal
             PET/fMRI, and hormonal assays are beginning to provide a
             mechanistic understanding of how individual differences in
             brain function arise. Against this background, functional
             genetic polymorphisms are being utilized to understand the
             origins of variability in signaling pathways as well as to
             efficiently model how such emergent variability impacts
             behaviorally relevant brain function and health outcomes.
             This chapter provides an overview of a research strategy
             that integrates these complimentary levels of analysis;
             existing empirical data is used to illustrate the
             effectiveness of this approach in illuminating the
             mechanistic neurobiology of individual differences in
             complex behavioral traits. This chapter also discusses how
             such efforts can contribute to the identification of
             predictive risk markers that interact with unique
             environmental factors to precipitate psychopathology.},
   Language = {eng},
   Doi = {10.1007/7854_2011_182},
   Key = {Bogdan2012c}
}

@article{Minkel2012,
   Author = {Minkel, JD and McNealy, K and Gianaros, PJ and Drabant, EM and Gross,
             JJ and Manuck, SB and Hariri, AR},
   Title = {Sleep quality and neural circuit function supporting emotion
             regulation},
   Journal = {Biology of mood \& anxiety disorders},
   Volume = {2},
   Number = {1},
   Pages = {22},
   Publisher = {Springer Science and Business Media LLC},
   Address = {Laboratory of NeuroGenetics, Department of Psychology \&
             Neuroscience, Duke University, 417 Chapel Dr,, Durham, NC
             27708, USA. jared.minkel@duke.edu.},
   Year = {2012},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23216889},
   Abstract = {ABSTRACT: BACKGROUND: Recent laboratory studies employing an
             extended sleep deprivation model have mapped sleep-related
             changes in behavior onto functional alterations in specific
             brain regions supporting emotion, suggesting possible
             biological mechanisms for an association between sleep
             difficulties and deficits in emotion regulation. However, it
             is not yet known if similar behavioral and neural changes
             are associated with the more modest variability in sleep
             observed in daily life. METHODS: We examined relationships
             between sleep and neural circuitry of emotion using the
             Pittsburgh Sleep Quality Index and fMRI data from a widely
             used emotion regulation task focusing on cognitive
             reappraisal of negative emotional stimuli in an unselected
             sample of 97 adult volunteers (48 women; mean age
             42.78+/-7.37years, range 30--54years old). RESULTS: Emotion
             regulation was associated with greater activation in
             clusters located in the dorsomedial prefrontal cortex
             (dmPFC), left dorsolateral prefrontal cortex (dlPFC), and
             inferior parietal cortex. Only one subscale from the
             Pittsburgh Sleep Quality Index, use of sleep medications,
             was related to BOLD responses in the dmPFC and dlPFC during
             cognitive reappraisal. Use of sleep medications predicted
             lesser BOLD responses during reappraisal, but other aspects
             of sleep, including sleep duration and subjective sleep
             quality, were not related to neural activation in this
             paradigm. CONCLUSIONS: The relatively modest variability in
             sleep that is common in the general community is unlikely to
             cause significant disruption in neural circuits supporting
             reactivity or regulation by cognitive reappraisal of
             negative emotion. Use of sleep medication however, may
             influence emotion regulation circuitry, but additional
             studies are necessary to determine if such use plays a
             causal role in altering emotional responses.},
   Language = {eng},
   Doi = {10.1186/2045-5380-2-22},
   Key = {Minkel2012}
}

@article{Carre2013b,
   Author = {Carre, JM and Hyde, LW and Neumann, CS and Viding, E and Hariri,
             AR},
   Title = {The neural signatures of distinct psychopathic
             traits},
   Journal = {Social neuroscience},
   Volume = {8},
   Number = {2},
   Pages = {122-135},
   Address = {Laboratory of NeuroGenetics, Department of Psychology and
             Neuroscience, Duke University, Durham, NC, USA.
             justin.carre@wayne.edu},
   Year = {2012},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22775289},
   Keywords = {Amygdala/pathology • Antisocial Personality
             Disorder/pathology/psychology • Data Interpretation,
             Statistical • Female • Humans • Image
             Processing, Computer-Assisted • Impulsive
             Behavior/pathology/psychology • Magnetic Resonance
             Imaging • Male • Neostriatum/pathology •
             Nervous System/*pathology • Neuropsychological Tests
             • Oxygen/blood • Reward • Sex Characteristics
             • Young Adult},
   Abstract = {Recent studies suggest that psychopathy may be associated
             with dysfunction in the neural circuitry supporting both
             threat- and reward-related processes. However, these studies
             have involved small samples and often focused on extreme
             groups. Thus, it is unclear to what extent current findings
             may generalize to psychopathic traits in the general
             population. Furthermore, no studies have systematically and
             simultaneously assessed associations between distinct
             psychopathy facets and both threat- and reward-related brain
             function in the same sample of participants. Here, we
             examined the relationship between threat-related amygdala
             reactivity and reward-related ventral striatum (VS)
             reactivity and variation in four facets of self-reported
             psychopathy in a sample of 200 young adults. Path models
             indicated that amygdala reactivity to fearful facial
             expressions is negatively associated with the interpersonal
             facet of psychopathy, whereas amygdala reactivity to angry
             facial expressions is positively associated with the
             lifestyle facet. Furthermore, these models revealed that
             differential VS reactivity to positive versus negative
             feedback is negatively associated with the lifestyle facet.
             There was suggestive evidence for gender-specific patterns
             of association between brain function and psychopathy
             facets. Our findings are the first to document differential
             associations between both threat- and reward-related neural
             processes and distinct facets of psychopathy and thus
             provide a more comprehensive picture of the pattern of
             neural vulnerabilities that may predispose to maladaptive
             outcomes associated with psychopathy.},
   Language = {eng},
   Doi = {10.1080/17470919.2012.703623},
   Key = {Carre2013b}
}

@article{Nikolova2012a,
   Author = {Nikolova, YS and Hariri, AR},
   Title = {Neural responses to threat and reward interact to predict
             stress-related problem drinking: A novel protective role of
             the amygdala},
   Journal = {Biology of mood \& anxiety disorders},
   Volume = {2},
   Number = {1},
   Pages = {19},
   Publisher = {Springer Science and Business Media LLC},
   Address = {Laboratory of NeuroGenetics, Department of Psychology \&
             Neuroscience and Institute for Genome Sciences \& Policy,
             Duke University, NC 27708, Durham, USA. yuliya.nikolova@duke.edu.},
   Year = {2012},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/23151390},
   Abstract = {ABSTRACT: BACKGROUND: Research into neural mechanisms of
             drug abuse risk has focused on the role of dysfunction in
             neural circuits for reward. In contrast, few studies have
             examined the role of dysfunction in neural circuits of
             threat in mediating drug abuse risk. Although typically
             regarded as a risk factor for mood and anxiety disorders,
             threat-related amygdala reactivity may serve as a protective
             factor against substance use disorders, particularly in
             individuals with exaggerated responsiveness to reward.
             FINDINGS: We used well-established neuroimaging paradigms to
             probe threat-related amygdala and reward-related ventral
             striatum reactivity in a sample of 200 young adult students
             from the ongoing Duke Neurogenetics Study. Recent life
             stress and problem drinking were assessed using self-report.
             We found a significant three-way interaction between
             threat-related amygdala reactivity, reward-related ventral
             striatum reactivity, and recent stress, wherein individuals
             with higher reward-related ventral striatum reactivity
             exhibit higher levels of problem drinking in the context of
             stress, but only if they also have lower threat-related
             amygdala reactivity. This three-way interaction predicted
             both contemporaneous problem drinking and problem drinking
             reported three-months later in a subset of participants.
             CONCLUSIONS: These findings suggest complex interactions
             between stress and neural responsiveness to both threat and
             reward mediate problem drinking. Furthermore, they highlight
             a novel protective role for threat-related amygdala
             reactivity against drug use in individuals with high neural
             reactivity to reward.},
   Language = {eng},
   Doi = {10.1186/2045-5380-2-19},
   Key = {Nikolova2012a}
}

@article{Gunduz-Cinar2013,
   Author = {Gunduz Cinar and O and Macpherson, KP and Cinar, R and Gamble George and J and Sugden, K and Williams, B and Godlewski, G and Ramikie, TS and Gorka,
             AX and Alapafuja, SO and Nikas, SP and Makriyannis, A and Poulton, R and Patel, S and Hariri, AR and Caspi, A and Moffitt, TE and Kunos, G and Holmes, A},
   Title = {Convergent translational evidence of a role for anandamide
             in amygdala-mediated fear extinction, threat processing and
             stress-reactivity},
   Journal = {Molecular psychiatry},
   Volume = {18},
   Number = {7},
   Pages = {813-823},
   Address = {Laboratory of Behavioral and Genomic Neuroscience, Section
             on Behavioral and Genomic Neuroscience, National Institute
             on Alcoholism and Alcohol Abuse, NIH, Bethesda, MD,
             USA.},
   Year = {2012},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22688188},
   Abstract = {Endocannabinoids are released 'on-demand' on the basis of
             physiological need, and can be pharmacologically augmented
             by inhibiting their catabolic degradation. The
             endocannabinoid anandamide is degraded by the catabolic
             enzyme fatty acid amide hydrolase (FAAH). Anandamide is
             implicated in the mediation of fear behaviors, including
             fear extinction, suggesting that selectively elevating brain
             anandamide could modulate plastic changes in fear. Here we
             first tested this hypothesis with preclinical experiments
             employing a novel, potent and selective FAAH inhibitor,
             AM3506 (5-(4-hydroxyphenyl)pentanesulfonyl fluoride).
             Systemic AM3506 administration before extinction decreased
             fear during a retrieval test in a mouse model of impaired
             extinction. AM3506 had no effects on fear in the absence of
             extinction training, or on various non-fear-related
             measures. Anandamide levels in the basolateral amygdala were
             increased by extinction training and augmented by systemic
             AM3506, whereas application of AM3506 to amygdala slices
             promoted long-term depression of inhibitory transmission, a
             form of synaptic plasticity linked to extinction. Further
             supporting the amygdala as effect-locus, the fear-reducing
             effects of systemic AM3506 were blocked by intra-amygdala
             infusion of a CB1 receptor antagonist and were fully
             recapitulated by intra-amygdala infusion of AM3506. On the
             basis of these preclinical findings, we hypothesized that
             variation in the human FAAH gene would predict individual
             differences in amygdala threat-processing and stress-coping
             traits. Consistent with this, carriers of a low-expressing
             FAAH variant (385A allele; rs324420) exhibited quicker
             habituation of amygdala reactivity to threat, and had lower
             scores on the personality trait of stress-reactivity. Our
             findings show that augmenting amygdala anandamide enables
             extinction-driven reductions in fear in mouse and may
             promote stress-coping in humans.Molecular Psychiatry advance
             online publication, 12 June 2012; doi:10.1038/mp.2012.72.},
   Language = {eng},
   Doi = {10.1038/mp.2012.72},
   Key = {Gunduz-Cinar2013}
}

@article{Lahey2012b,
   Author = {Lahey, BB and Applegate, B and Hakes, JK and Zald, DH and Hariri, AR and Rathouz, PJ},
   Title = {Is there a general factor of prevalent psychopathology
             during adulthood?},
   Journal = {Journal of abnormal psychology},
   Volume = {121},
   Number = {4},
   Pages = {971-977},
   Address = {Department of Health Studies (MC 2007), University of
             Chicago, Chicago, IL 60637, USA. blahey@uchicago.edu},
   Year = {2012},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22845652},
   Keywords = {Adolescent • Adult • Comorbidity • Diagnostic
             and Statistical Manual of Mental Disorders • Factor
             Analysis, Statistical • Female • Humans •
             Interview, Psychological • Male • Mental
             Disorders/diagnosis/*epidemiology • Middle Aged •
             *Models, Psychological • Prevalence},
   Abstract = {The patterns of comorbidity among prevalent mental disorders
             in adults lead them to load on ``externalizing,''
             ``distress,'' and ``fears'' factors. These factors are
             themselves robustly correlated, but little attention has
             been paid to this fact. As a first step in studying the
             implications of these interfactor correlations, we conducted
             confirmatory factor analyses on diagnoses of 11 prevalent
             Diagnostic and Statistical Manual of Mental Disorders (4th
             ed.) mental disorders in a nationally representative sample.
             A model specifying correlated externalizing, distress, and
             fears factors fit well, but an alternative model was tested
             in which a ``general'' bifactor was added to capture what
             these disorders share in common. There was a modest but
             significant improvement in fit for the bifactor model
             relative to the 3-factor oblique model, with all disorders
             loading strongly on the bifactor. Tests of external validity
             revealed that the fears, distress, and externalizing factors
             were differentially associated with measures of functioning
             and potential risk factors. Nonetheless, the general
             bifactor accounted for significant independent variance in
             future psychopathology, functioning, and other criteria over
             and above the fears, distress, and externalizing factors.
             These findings support the hypothesis that these prevalent
             forms of psychopathology have both important common and
             unique features. Future studies should determine whether
             this is because they share elements of their etiology and
             neurobiological mechanisms. If so, the existence of common
             features across diverse forms of prevalent psychopathology
             could have important implications for understanding the
             nature, etiology, and outcomes of psychopathology. (PsycINFO
             Database Record (c) 2012 APA, all rights
             reserved).},
   Language = {eng},
   Doi = {10.1037/a0028355},
   Key = {Lahey2012b}
}

@article{Hyde2011b,
   Author = {Hyde, LW and Bogdan, R and Hariri, AR},
   Title = {Understanding risk for psychopathology through imaging
             gene-environment interactions.},
   Journal = {Trends in cognitive sciences},
   Volume = {15},
   Number = {9},
   Pages = {417-427},
   Address = {Department of Psychology and Center for the Neural Basis of
             Cognition, University of Pittsburgh, 210 South Bouquet St,
             Pittsburgh, PA 15260, USA. LWH2@pitt.edu},
   Year = {2011},
   Month = {September},
   ISSN = {1364-6613},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21839667},
   Keywords = {Brain/*pathology/physiopathology • Epigenomics •
             *Gene-Environment Interaction • *Genetic Predisposition
             to Disease • Humans • Models, Biological •
             Neuroimaging • *Psychopathology},
   Abstract = {Examining the interplay of genes, experience and the brain
             is crucial to understanding psychopathology. We review the
             recent gene-environment interaction (G×E) and imaging
             genetics literature with the goal of developing models to
             bridge these approaches within single imaging
             gene-environment interaction (IG×E) studies. We explore
             challenges inherent in both G×E and imaging genetics and
             highlight studies that address these limitations. In
             specifying IG×E models, we examine statistical methods for
             combining these approaches, and explore plausible biological
             mechanisms (e.g. epigenetics) through which these
             conditional mechanisms can be understood. Finally, we
             discuss the potential contribution that IG×E studies can
             make to understanding psychopathology and developing more
             personalized and effective prevention and
             treatment.},
   Language = {eng},
   Doi = {10.1016/j.tics.2011.07.001},
   Key = {Hyde2011b}
}

@article{Nikolova2011,
   Author = {Nikolova, YS and Ferrell, RE and Manuck, SB and Hariri,
             AR},
   Title = {Multilocus genetic profile for dopamine signaling predicts
             ventral striatum reactivity.},
   Journal = {Neuropsychopharmacology : official publication of the
             American College of Neuropsychopharmacology},
   Volume = {36},
   Number = {9},
   Pages = {1940-1947},
   Address = {Department of Psychology and Neuroscience, Duke University,
             Durham, NC, USA.},
   Year = {2011},
   Month = {August},
   ISSN = {0893-133X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21593733},
   Keywords = {Adult • Basal Ganglia/*metabolism/physiopathology
             • Cohort Studies • Dopamine/genetics/*physiology
             • Dopamine Plasma Membrane Transport Proteins/genetics
             • Female • Gene Expression Profiling/*methods
             • Genetic Loci/*genetics • Genetic Predisposition
             to Disease/genetics • Humans • Male • Middle
             Aged • Mood Disorders/diagnosis/*genetics/physiopathology
             • Polymorphism, Genetic/genetics • Predictive
             Value of Tests • Receptors, Dopamine D2/genetics •
             Receptors, Dopamine D4/genetics • Signal
             Transduction/*genetics},
   Abstract = {Research integrating neuroimaging and molecular genetics has
             yielded important insights into how variability in brain
             chemistry predicts individual differences in brain function,
             behavior and related risk for psychopathology. However,
             existing studies have been limited by their focus on the
             independent effects of single polymorphisms with modest
             impact on brain chemistry. Here, we explored the effects of
             five functional polymorphisms affecting dopamine (DA)
             signaling on reward-related ventral striatum (VS)
             reactivity, measured with BOLD fMRI, in a sample of 69
             Caucasians. We also compiled individual multilocus genetic
             profile scores reflecting the additive effects of alleles
             conferring relatively increased DA signaling across the five
             polymorphic loci: DAT1 9-repeat, DRD4 7-repeat, DRD2 -141C
             Del, DRD2 Taq1A C (A2), and COMT (158)Met. These multilocus
             DA profile scores accounted for 10.9% of the
             inter-individual variability in reward-related VS
             reactivity. In contrast, none of the individual
             polymorphisms accounted for significant variability. Our
             results show that biologically informed multilocus genetic
             profiles have unique promise as indices of variability in
             brain chemistry that may yield advances in mapping
             individual differences in behaviorally relevant brain
             function. In turn, such genetic profiles may fuel
             gene-environment interactions research establishing
             trajectories of risk for psychopathology.},
   Language = {eng},
   Doi = {10.1038/npp.2011.82},
   Key = {Nikolova2011}
}

@article{Carre2011,
   Author = {Carré, JM and McCormick, CM and Hariri, AR},
   Title = {The social neuroendocrinology of human aggression.},
   Journal = {Psychoneuroendocrinology},
   Volume = {36},
   Number = {7},
   Pages = {935-944},
   Address = {Department of Psychology and Neuroscience, Duke University,
             Durham, NC, USA. justin.carre@duke.edu},
   Year = {2011},
   Month = {August},
   ISSN = {0306-4530},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21367531},
   Keywords = {Aggression/*physiology/psychology • Animals •
             Competitive Behavior/physiology • Humans • Models,
             Animal • Models, Biological • Neural
             Pathways/metabolism/physiology • Neuroendocrinology
             • Neurosecretory Systems/metabolism/*physiology •
             *Social Behavior • Testosterone/blood/physiology},
   Abstract = {Testosterone concentrations fluctuate rapidly in response to
             competitive and aggressive interactions, suggesting that
             changes in testosterone rather than baseline differences
             shape ongoing and/or future competitive and aggressive
             behaviors. Although recent experiments in animal models
             provide compelling empirical support for this idea, studies
             in humans have focused largely on how competitive
             interactions drive changes in testosterone concentrations
             and not how these changes influence subsequent behavior. In
             this paper, we provide a review of the literature on
             testosterone and human aggression with a main focus on the
             role of testosterone dynamics in modulating reactive
             aggression. We also speculate on one putative neural
             mechanism through which testosterone may bias human
             aggressive behavior. Finally, we conclude by highlighting
             important questions that should be addressed in future
             research.},
   Language = {eng},
   Doi = {10.1016/j.psyneuen.2011.02.001},
   Key = {Carre2011}
}

@article{Hariri2011c,
   Author = {Hariri, AR},
   Title = {The what, where, and when of catechol-O-methyltransferase.},
   Journal = {Biological psychiatry},
   Volume = {70},
   Number = {3},
   Pages = {214-215},
   Address = {Laboratory of NeuroGenetics, Department of Psychology and
             Neuroscience, Institute for Genome Sciences and Policy, Duke
             University, 417 Chapel Drive, Durham, NC 27708, USA.
             ahmad.hariri@duke.edu},
   Year = {2011},
   Month = {August},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21745617},
   Keywords = {Attention Deficit Disorder with Hyperactivity/*genetics
             • Brain/*physiology • Catechol
             O-Methyltransferase/*genetics • Central Nervous System
             Stimulants/*therapeutic use • Female • Humans
             • Male • Memory, Short-Term/*physiology •
             Methylphenidate/*therapeutic use • *Polymorphism,
             Single Nucleotide},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2011.06.002},
   Key = {Hariri2011c}
}

@article{Morey2011,
   Author = {Morey, RA and Hariri, AR and Gold, AL and Hauser, MA and Munger, HJ and Dolcos, F and McCarthy, G},
   Title = {Serotonin transporter gene polymorphisms and brain function
             during emotional distraction from cognitive processing in
             posttraumatic stress disorder.},
   Journal = {BMC Psychiatry},
   Volume = {11},
   Pages = {76},
   Address = {Department of Psychiatry and Behavioral Sciences, Duke
             University, Durham, NC 27710, USA. morey@biac.duke.edu},
   Year = {2011},
   Month = {May},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21545724},
   Keywords = {Amygdala/physiopathology • Case-Control Studies •
             Cognition/*physiology • Emotions/*physiology •
             Functional Neuroimaging/methods/psychology • Humans
             • Magnetic Resonance Imaging/psychology • Memory,
             Short-Term/physiology • *Polymorphism, Single
             Nucleotide • Prefrontal Cortex/physiopathology •
             Promoter Regions, Genetic/genetics/physiology •
             Psychomotor Performance/physiology • September 11
             Terrorist Attacks/psychology • Serotonin Plasma
             Membrane Transport Proteins/genetics/*physiology •
             Stress Disorders, Post-Traumatic/*genetics/*physiopathology/psychology
             • Veterans/psychology • Wounds and
             Injuries/physiopathology/psychology},
   Abstract = {BACKGROUND: Serotonergic system dysfunction has been
             implicated in posttraumatic stress disorder (PTSD). Genetic
             polymorphisms associated with serotonin signaling may
             predict differences in brain circuitry involved in emotion
             processing and deficits associated with PTSD. In healthy
             individuals, common functional polymorphisms in the
             serotonin transporter gene (SLC6A4) have been shown to
             modulate amygdala and prefrontal cortex (PFC) activity in
             response to salient emotional stimuli. Similar patterns of
             differential neural responses to emotional stimuli have been
             demonstrated in PTSD but genetic factors influencing these
             activations have yet to be examined. METHODS: We
             investigated whether SLC6A4 promoter polymorphisms
             (5-HTTLPR, rs25531) and several downstream single nucleotide
             polymorphisms (SNPs) modulated activity of brain regions
             involved in the cognitive control of emotion in post-9/11
             veterans with PTSD. We used functional MRI to examine neural
             activity in a PTSD group (n = 22) and a trauma-exposed
             control group (n = 20) in response to trauma-related images
             presented as task-irrelevant distractors during the active
             maintenance period of a delayed-response working memory
             task. Regions of interest were derived by contrasting
             activation for the most distracting and least distracting
             conditions across participants. RESULTS: In patients with
             PTSD, when compared to trauma-exposed controls, rs16965628
             (associated with serotonin transporter gene expression)
             modulated task-related ventrolateral PFC activation and
             5-HTTLPR tended to modulate left amygdala activation.
             Subsequent to combat-related trauma, these SLC6A4
             polymorphisms may bias serotonin signaling and the neural
             circuitry mediating cognitive control of emotion in patients
             with PTSD. CONCLUSIONS: The SLC6A4 SNP rs16965628 and
             5-HTTLPR are associated with a bias in neural responses to
             traumatic reminders and cognitive control of emotions in
             patients with PTSD. Functional MRI may help identify
             intermediate phenotypes and dimensions of PTSD that clarify
             the functional link between genes and disease phenotype, and
             also highlight features of PTSD that show more proximal
             influence of susceptibility genes compared to current
             clinical categorizations.},
   Language = {eng},
   Doi = {10.1186/1471-244X-11-76},
   Key = {Morey2011}
}

@article{Gianaros2011,
   Author = {Gianaros, PJ and Manuck, SB and Sheu, LK and Kuan, DCH and Votruba-Drzal, E and Craig, AE and Hariri, AR},
   Title = {Parental education predicts corticostriatal functionality in
             adulthood.},
   Journal = {Cerebral cortex (New York, N.Y. : 1991)},
   Volume = {21},
   Number = {4},
   Pages = {896-910},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, PA 15213, USA. gianarospj@upmc.edu},
   Year = {2011},
   Month = {April},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20810623},
   Keywords = {Adult • Cerebral Cortex/*physiology • *Educational
             Status • Female • Humans • Magnetic Resonance
             Imaging • Male • Middle Aged • Neural
             Pathways/*physiology • *Parents • Socioeconomic
             Factors},
   Abstract = {Socioeconomic disadvantage experienced in early development
             predicts ill health in adulthood. However, the
             neurobiological pathways linking early disadvantage to adult
             health remain unclear. Lower parental education-a
             presumptive indicator of early socioeconomic
             disadvantage-predicts health-impairing adult behaviors,
             including tobacco and alcohol dependencies. These behaviors
             depend, in part, on the functionality of corticostriatal
             brain systems that 1) show developmental plasticity and
             early vulnerability, 2) process reward-related information,
             and 3) regulate impulsive decisions and actions. Hence,
             corticostriatal functionality in adulthood may covary
             directly with indicators of early socioeconomic
             disadvantage, particularly lower parental education. Here,
             we tested the covariation between parental education and
             corticostriatal activation and connectivity in 76 adults
             without confounding clinical syndromes. Corticostriatal
             activation and connectivity were assessed during the
             processing of stimuli signaling monetary gains (positive
             feedback [PF]) and losses (negative feedback). After
             accounting for participants' own education and other
             explanatory factors, lower parental education predicted
             reduced activation in anterior cingulate and dorsomedial
             prefrontal cortices during PF, along with reduced
             connectivity between these cortices and orbitofrontal and
             striatal areas implicated in reward processing and impulse
             regulation. In speculation, adult alterations in
             corticostriatal functionality may represent facets of a
             neurobiological endophenotype linked to socioeconomic
             conditions of early development.},
   Language = {eng},
   Doi = {10.1093/cercor/bhq160},
   Key = {Gianaros2011}
}

@article{Drabant2011,
   Author = {Drabant, EM and Kuo, JR and Ramel, W and Blechert, J and Edge, MD and Cooper, JR and Goldin, PR and Hariri, AR and Gross,
             JJ},
   Title = {Experiential, autonomic, and neural responses during threat
             anticipation vary as a function of threat intensity and
             neuroticism.},
   Journal = {NeuroImage},
   Volume = {55},
   Number = {1},
   Pages = {401-410},
   Address = {Psychology Department, Stanford University, Stanford, CA
             94305, USA. drabant@stanford.edu},
   Year = {2011},
   Month = {March},
   ISSN = {1053-8119},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21093595},
   Keywords = {Brain/*physiopathology • *Fear • Female •
             Humans • *Magnetic Resonance Imaging • Neurotic
             Disorders/*physiopathology • *Personal Autonomy •
             *Problem-Based Learning • Young Adult},
   Abstract = {Anticipatory emotional responses play a crucial role in
             preparing individuals for impending challenges. They do this
             by triggering a coordinated set of changes in behavioral,
             autonomic, and neural response systems. In the present
             study, we examined the biobehavioral impact of varying
             levels of anticipatory anxiety, using a shock anticipation
             task in which unpredictable electric shocks were threatened
             and delivered to the wrist at variable intervals and
             intensities (safe, medium, strong). This permitted
             investigation of a dynamic range of anticipatory anxiety
             responses. In two studies, 95 and 51 healthy female
             participants, respectively, underwent this shock
             anticipation task while providing continuous ratings of
             anxiety experience and electrodermal responding (Study 1)
             and during fMRI BOLD neuroimaging (Study 2). Results
             indicated a step-wise pattern of responding in anxiety
             experience and electrodermal responses. Several brain
             regions showed robust responses to shock anticipation
             relative to safe trials, including the hypothalamus,
             periaqueductal gray, caudate, precentral gyrus, thalamus,
             insula, ventrolateral PFC, dorsomedial PFC, and ACC. A
             subset of these regions demonstrated a linear pattern of
             increased responding from safe to medium to strong trials,
             including the bilateral insula, ACC, and inferior frontal
             gyrus. These responses were modulated by individual
             differences in neuroticism, such that those high in
             neuroticism showed exaggerated anxiety experience across the
             entire task, and reduced brain activation from medium to
             strong trials in a subset of brain regions. These findings
             suggest that individual differences in neuroticism may
             influence sensitivity to anticipatory threat and provide new
             insights into the mechanism through which neuroticism may
             confer risk for developing anxiety disorders via
             dysregulated anticipatory responses.},
   Language = {eng},
   Doi = {10.1016/j.neuroimage.2010.11.040},
   Key = {Drabant2011}
}

@article{Hyde2011a,
   Author = {Hyde, LW and Gorka, A and Manuck, SB and Hariri, AR},
   Title = {Perceived social support moderates the link between
             threat-related amygdala reactivity and trait
             anxiety.},
   Journal = {Neuropsychologia},
   Volume = {49},
   Number = {4},
   Pages = {651-656},
   Address = {Department of Psychology, University of Pittsburgh,
             Pittsburgh, PA 15260, USA. lwh2@pitt.edu},
   Year = {2011},
   Month = {March},
   ISSN = {0028-3932},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20813118},
   Keywords = {*Adaptation, Psychological • Adult •
             Aggression/*psychology • Amygdala/*physiology •
             Anxiety/*psychology • Brain Mapping • Female
             • Humans • Magnetic Resonance Imaging • Male
             • Middle Aged • Reference Values • *Social
             Support • Temperament},
   Abstract = {Several lines of research have illustrated that negative
             environments can precipitate psychopathology, particularly
             in the context of relatively increased biological risk,
             while social resources can buffer the effects of these
             environments. However, little research has examined how
             social resources might buffer proximal biological risk for
             psychopathology or the neurobiological pathways through
             which such buffering may be mediated. Here we report that
             the expression of trait anxiety as a function of
             threat-related amygdala reactivity is moderated by perceived
             social support, a resource for coping with adversity. A
             significant positive correlation between amygdala reactivity
             and trait anxiety was evident in individuals reporting below
             average levels of support but not in those reporting average
             or above average levels. These results were consistent
             across multiple measures of trait anxiety and were specific
             to anxiety in that they did not extend to measures of broad
             negative or positive affect. Our findings illuminate a
             biological pathway, namely moderation of amygdala-related
             anxiety, through which social support may confer resilience
             to psychopathology. Moreover, our results indicate that
             links between neural reactivity and behavior are not static
             but rather may be contingent on social resources.},
   Language = {eng},
   Doi = {10.1016/j.neuropsychologia.2010.08.025},
   Key = {Hyde2011a}
}

@article{Hariri2011a,
   Author = {Hariri, AR and Whalen, PJ},
   Title = {The amygdala: Inside and out},
   Journal = {F1000 Biology Reports},
   Volume = {3},
   Number = {1},
   Pages = {2},
   Publisher = {Faculty of 1000, Ltd.},
   Year = {2011},
   Month = {February},
   ISSN = {1740-4118},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21399763},
   Abstract = {Research at the interface of psychology, neuroscience,
             molecular biology, and genetics, focusing on the amygdala,
             has begun to reveal a rule book for emotional reactions.
             Variations in intrinsic and extrinsic factors tweak the
             sensitivity of the amygdala, giving rise to differences in
             behavior between individuals. At their most extreme, these
             variations may generate psychological disorders, and even
             our current rudimentary understanding of this brain region
             suggests novel strategies for the treatment of such
             disorders. © 2011 Faculty of 1000 Ltd.},
   Language = {eng},
   Doi = {10.3410/B3-2},
   Key = {Hariri2011a}
}

@article{fds252004,
   Author = {Hariri, AR and Whalen, PJ},
   Title = {Face to face with the emotional brain},
   Journal = {Scientist},
   Volume = {25},
   Number = {2},
   Pages = {30},
   Year = {2011},
   Month = {February},
   ISSN = {0890-3670},
   Key = {fds252004}
}

@article{Beevers2011,
   Author = {Beevers, CG and Marti, CN and Lee, H-J and Stote, DL and Ferrell, RE and Hariri, AR and Telch, MJ},
   Title = {Associations between serotonin transporter gene promoter
             region (5-HTTLPR) polymorphism and gaze bias for emotional
             information.},
   Journal = {Journal of abnormal psychology},
   Volume = {120},
   Number = {1},
   Pages = {187-197},
   Address = {Department of Psychology, University of Texas at Austin, 1
             University Station, A8000, Austin, TX 78712, USA.
             beevers@psy.utexas.edu},
   Year = {2011},
   Month = {February},
   ISSN = {0021-843X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/21319930},
   Keywords = {Adult • Alleles • Analysis of Variance •
             Attention/*physiology • Emotions/*physiology • Eye
             Movements/*genetics • Facial Expression • Female
             • Gene Frequency • Genotype • Humans •
             Male • Photic Stimulation • *Polymorphism, Single
             Nucleotide • Promoter Regions, Genetic • Serotonin
             Plasma Membrane Transport Proteins/*genetics • Time
             Factors},
   Abstract = {The serotonin transporter promoter region polymorphism
             (5-HTTLPR) is associated with neural response to negative
             images in brain regions involved in the experience of
             emotion. However, the behavioral implications of this
             sensitivity have been studied far less extensively. The
             current study used eye-tracking methodology to examine how
             individuals genotyped for the 5-HTTLPR, including the single
             nucleotide polymorphism (SNP) rs25531, allocated attention
             during prolonged (30-s) exposure to face stimuli depicting
             positive and negative emotion. Short 5-HTTLPR allele
             carriers and carriers of the long allele with guanine at the
             sixth nucleotide (S/LG) displayed a stronger gaze bias
             (total fixation time, number of fixations, mean fixation
             length) for positive than for sad, threat, or neutral
             stimuli. In contrast, those homozygous for the long 5-HTTLPR
             allele with adenine at the sixth nucleotide (LA) viewed the
             emotion stimuli in an unbiased fashion. Time course analyses
             indicated no initial 5-HTTLPR group differences; however,
             S/LG 5-HTTLPR allele carriers were more likely than LA
             5-HTTLPR homozygotes to direct gaze toward happy than toward
             sad stimuli over time. This bias toward positive stimuli
             during the later stages of information processing likely
             reflects a strategic effort to downregulate heightened
             reactivity to negative stimuli among 5-HTTLPR S/LG allele
             carriers.},
   Language = {eng},
   Doi = {10.1037/a0022125},
   Key = {Beevers2011}
}

@article{Hariri2011b,
   Author = {Hariri, AR and Shin, LM},
   Title = {Welcome to biology of mood \& anxiety disorders},
   Journal = {Biology of mood \& anxiety disorders},
   Volume = {1},
   Number = {1},
   Pages = {1},
   Publisher = {Springer Science and Business Media LLC},
   Address = {Laboratory of NeuroGenetics, Department of Psychology \&
             Neuroscience, Institute for Genome Sciences \& Policy, Duke
             University, Durham, NC USA. ahmad.hariri@duke.edu.},
   Year = {2011},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22738418},
   Language = {eng},
   Doi = {10.1186/2045-5380-1-1},
   Key = {Hariri2011b}
}

@article{fds252110,
   Author = {Carre, JM and Murphy, KR and Hariri, AR},
   Title = {What lies beneath the face of aggression?},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {8},
   Number = {2},
   Pages = {224-229},
   Year = {2011},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22198969},
   Abstract = {Recent evidence indicates that a sexually dimorphic feature
             of humans, the facial width-to-height ratio (FWHR), is
             positively correlated with reactive aggression, particularly
             in men. Also, predictions about the aggressive tendencies of
             others faithfully map onto FWHR in the absence of explicit
             awareness of this metric. Here, we provide the first
             evidence that amygdala reactivity to social signals of
             interpersonal challenge may underlie the link between
             aggression and the FWHR. Specifically, amygdala reactivity
             to angry faces was positively correlated with aggression,
             but only among men with relatively large FWHRs. The patterns
             of association were specific to angry facial expressions and
             unique to men. These links may reflect the common influence
             of pubertal testosterone on craniofacial growth and
             development of neural circuitry underlying aggression.
             Amygdala reactivity may also represent a plausible pathway
             through which FWHR may have evolved to represent an honest
             indicator of conspecific threat, namely by reflecting the
             responsiveness of neural circuitry mediating aggressive
             behavior.},
   Doi = {10.1093/scan/nsr096},
   Key = {fds252110}
}

@article{Fisher2011,
   Author = {Fisher, PM and Price, JC and Meltzer, CC and Moses Kolko and EL and Becker,
             C and Berga, SL and Hariri, AR},
   Title = {Medial prefrontal cortex serotonin 1A and 2A receptor
             binding interacts to predict threat-related amygdala
             reactivity},
   Journal = {Biology of mood \& anxiety disorders},
   Volume = {1},
   Number = {1},
   Pages = {2},
   Publisher = {Springer Science and Business Media LLC},
   Address = {Center for Neuroscience, University of Pittsburgh,
             Pittsburgh, Pennsylvania 15260, USA. patrick.fisher@gmail.com.},
   Year = {2011},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/22738071},
   Abstract = {BACKGROUND: The amygdala and medial prefrontal cortex (mPFC)
             comprise a key corticolimbic circuit that helps shape
             individual differences in sensitivity to threat and the
             related risk for psychopathology. Although serotonin (5-HT)
             is known to be a key modulator of this circuit, the specific
             receptors mediating this modulation are unclear. The
             colocalization of 5-HT1A and 5-HT2A receptors on mPFC
             glutamatergic neurons suggests that their functional
             interactions may mediate 5-HT effects on this circuit
             through top-down regulation of amygdala reactivity. Using a
             multimodal neuroimaging strategy in 39 healthy volunteers,
             we determined whether threat-related amygdala reactivity,
             assessed with blood oxygen level-dependent functional
             magnetic resonance imaging, was significantly predicted by
             the interaction between mPFC 5-HT1A and 5-HT2A receptor
             levels, assessed by positron emission tomography. RESULTS:
             5-HT1A binding in the mPFC significantly moderated an
             inverse correlation between mPFC 5-HT2A binding and
             threat-related amygdala reactivity. Specifically, mPFC
             5-HT2A binding was significantly inversely correlated with
             amygdala reactivity only when mPFC 5-HT1A binding was
             relatively low. CONCLUSIONS: Our findings provide evidence
             that 5-HT1A and 5-HT2A receptors interact to shape
             serotonergic modulation of a functional circuit between the
             amygdala and mPFC. The effect of the interaction between
             mPFC 5-HT1A and 5-HT2A binding and amygdala reactivity is
             consistent with the colocalization of these receptors on
             glutamatergic neurons in the mPFC.},
   Language = {eng},
   Doi = {10.1186/2045-5380-1-2},
   Key = {Fisher2011}
}

@article{Cornelius2010,
   Author = {Cornelius, JR and Aizenstein, HJ and Hariri, AR},
   Title = {Amygdala reactivity is inversely related to level of
             cannabis use in individuals with comorbid cannabis
             dependence and major depression.},
   Journal = {Addictive behaviors},
   Volume = {35},
   Number = {6},
   Pages = {644-646},
   Address = {Department of Psychiatry, University of Pittsburgh School of
             Medicine, Pittsburgh, PA 15213, United States.
             corneliusjr@upmc.edu},
   Year = {2010},
   Month = {June},
   ISSN = {0306-4603},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20189314},
   Keywords = {Amygdala/*drug effects/physiopathology • Antidepressive
             Agents, Second-Generation/therapeutic use •
             Cannabis/*adverse effects • Comorbidity •
             Depressive Disorder/drug therapy/epidemiology •
             Diagnosis, Dual (Psychiatry) • Double-Blind Method
             • Fear/*drug effects • Female •
             Fluoxetine/therapeutic use • Humans • Magnetic
             Resonance Imaging/methods • Male • Marijuana
             Abuse/epidemiology/*physiopathology • Social Behavior
             • Young Adult},
   Abstract = {Phan et al. (2008) recently reported that an acute dose of
             oral THC is associated with a decrease in threat-related
             amygdala reactivity during a social threat stimulus task.
             However, to date, those findings have not been replicated,
             and have not been extended to clinical studies involving
             smoked rather than oral cannabis. In this study, we
             hypothesized that level of cannabis smoked by participants
             in our treatment study would be inversely related to the
             level of threat-related amygdala reactivity. Subjects were
             recruited from among participants in our double-blind,
             placebo-controlled trial of fluoxetine in comorbid youth
             with cannabis dependence/major depression. The
             threat-related amygdala reactivity task used by Hariri et
             al. (2009) was completed during BOLD fMRI scans at study
             baseline and then again 12 weeks later at the end of the
             trial. Data are available from six subjects with pre-and
             post-treatment fMRI data. During the course of the study,
             five of the six subjects demonstrated a decrease in their
             level of cannabis use, with a mean decrease of 64%, and
             those persons all demonstrated an increase in their level of
             amygdala reactivity. One subject demonstrated an increase in
             their level of cannabis use (a 79% increase) during the
             treatment trial, and that person demonstrated a decrease in
             their level of amygdala reactivity. Thus, a higher level of
             cannabis use was consistently associated with a lower level
             of amygdala reactivity across all subjects (matched pairs t
             = 2.70, df = 5, p < 0.05, two-tailed). These findings are
             consistent with the reports by Phan et al. (2008) and Hariri
             et al. (2009) suggesting that cannabinoids have an
             inhibitory effect on threat-related amygdala
             reactivity.},
   Language = {eng},
   Doi = {10.1016/j.addbeh.2010.02.004},
   Key = {Cornelius2010}
}

@article{Chiao2010,
   Author = {Chiao, JY and Hariri, AR and Harada, T and Mano, Y and Sadato, N and Parrish, TB and Iidaka, T},
   Title = {Theory and methods in cultural neuroscience.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {5},
   Number = {2-3},
   Pages = {356-361},
   Address = {Department of Psychology and Interdepartmental Neuroscience
             Program, Northwestern University, Evanston, IL 60208, USA.
             jchiao@northwestern.edu},
   Year = {2010},
   Month = {June},
   ISSN = {1749-5016},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20592044},
   Keywords = {Brain/physiology • Brain Mapping • Cross-Cultural
             Comparison • *Culture • Humans • Magnetic
             Resonance Imaging • Neurosciences/*methods •
             Polymorphism, Genetic/genetics • Research Design •
             Stereotyping},
   Abstract = {Cultural neuroscience is an emerging research discipline
             that investigates cultural variation in psychological,
             neural and genomic processes as a means of articulating the
             bidirectional relationship of these processes and their
             emergent properties. Research in cultural neuroscience
             integrates theory and methods from anthropology, cultural
             psychology, neuroscience and neurogenetics. Here, we review
             a set of core theoretical and methodological challenges
             facing researchers when planning and conducting cultural
             neuroscience studies, and provide suggestions for overcoming
             these challenges. In particular, we focus on the problems of
             defining culture and culturally appropriate experimental
             tasks, comparing neuroimaging data acquired from different
             populations and scanner sites and identifying functional
             genetic polymorphisms relevant to culture. Implications of
             cultural neuroscience research for addressing current issues
             in population health disparities are discussed.},
   Language = {eng},
   Doi = {10.1093/scan/nsq063},
   Key = {Chiao2010}
}

@article{fds252003,
   Author = {Jedema, HP and Gianaros, PJ and Greer, PJ and Kerr, DD and Liu, S and Higley, JD and Suomi, SJ and Olsen, AS and Porter, JN and Lopresti, BJ and Hariri, AR and Bradberry, CW},
   Title = {Morphological differences associated with a serotonin
             transporter polymorphism (5-HTTLPR) in rhesus
             macaques},
   Journal = {Molecular Psychiatry},
   Volume = {15},
   Number = {5},
   Pages = {446},
   Publisher = {Springer Nature},
   Year = {2010},
   Month = {May},
   ISSN = {1359-4184},
   Doi = {10.1038/mp.2010.49},
   Key = {fds252003}
}

@article{Jedema2010,
   Author = {Jedema, HP and Gianaros, PJ and Greer, PJ and Kerr, DD and Liu, S and Higley, JD and Suomi, SJ and Olsen, AS and Porter, JN and Lopresti, BJ and Hariri, AR and Bradberry, CW},
   Title = {Cognitive impact of genetic variation of the serotonin
             transporter in primates is associated with differences in
             brain morphology rather than serotonin neurotransmission.},
   Journal = {Molecular psychiatry},
   Volume = {15},
   Number = {5},
   Pages = {512-446},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, PA 15261, USA.},
   Year = {2010},
   Month = {May},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19721434},
   Keywords = {Animals • Avoidance Learning/physiology •
             Behavior, Animal/physiology • Benzylamines/metabolism
             • Brain/drug effects/radionuclide imaging • Brain
             Mapping • Carbon Isotopes/metabolism • Choice
             Behavior/*physiology • Cognition/*physiology •
             Genotype • Macaca mulatta • Magnetic Resonance
             Imaging/methods • Male • Neuropsychological Tests
             • Piperazines/metabolism • Polymorphism,
             Genetic/*genetics • Positron-Emission
             Tomography/methods • Protein Binding/drug
             effects/genetics • Pyridines/metabolism •
             Receptor, Serotonin, 5-HT1A/genetics •
             Serotonin/genetics/*metabolism • Serotonin Plasma
             Membrane Transport Proteins/*genetics • Synaptic
             Transmission/*genetics • Time Factors •
             Tritium/metabolism},
   Abstract = {A powerful convergence of genetics, neuroimaging and
             epidemiological research has identified the biological
             pathways mediating individual differences in complex
             behavioral processes and the related risk for disease.
             Orthologous genetic variation in non-human primates (NHPs)
             represents a unique opportunity to characterize the detailed
             molecular and cellular mechanisms that bias behaviorally and
             clinically relevant brain function. We report that a rhesus
             macaque orthologue of a common polymorphism of the serotonin
             transporter gene (rh5-HTTLPR) has strikingly similar effects
             on behavior and brain morphology to those in humans.
             Specifically, the rh5-HTTLPR (S)hort allele broadly affects
             cognitive choice behavior and brain morphology without
             observably affecting the 5-hydroxytryptamine (5-HT)
             transporter or 5-HT(1A) concentrations in vivo.
             Collectively, our findings indicate that 5-HTTLPR-associated
             behavioral effects reflect genotype-dependent biases in
             cortical development rather than static differences in
             serotonergic signaling mechanisms. Moreover, these data
             highlight the vast potential of NHP models in advancing our
             understanding of human genetic variation affecting behavior
             and neuropsychiatric disease liability.},
   Language = {eng},
   Doi = {10.1038/mp.2009.90},
   Key = {Jedema2010}
}

@article{Caspi2010,
   Author = {Caspi, A and Hariri, AR and Holmes, A and Uher, R and Moffitt,
             TE},
   Title = {Genetic sensitivity to the environment: the case of the
             serotonin transporter gene and its implications for studying
             complex diseases and traits.},
   Journal = {The American journal of psychiatry},
   Volume = {167},
   Number = {5},
   Pages = {509-527},
   Address = {Department of Psychology, and Institute for Genome Sciences
             and Policy, Duke University, Durham, NC 27708, USA.
             ac115@duke.edu},
   Year = {2010},
   Month = {May},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20231323},
   Keywords = {Animals • Brain/pathology/physiopathology •
             Depressive Disorder/genetics • Environment •
             Genetic Predisposition to Disease/*genetics •
             Genome-Wide Association Study/methods • Genotype •
             Humans • Models, Psychological • Mood
             Disorders/genetics • Primates/genetics/psychology
             • Serotonin Plasma Membrane Transport
             Proteins/*genetics/physiology • Stress,
             Psychological/complications/*genetics},
   Abstract = {Evidence of marked variability in response among people
             exposed to the same environmental risk implies that
             individual differences in genetic susceptibility might be at
             work. The study of such Gene-by-Environment (GxE)
             interactions has gained momentum. In this article, the
             authors review research about one of the most extensive
             areas of inquiry: variation in the promoter region of the
             serotonin transporter gene (SLC6A4; also known as 5-HTT) and
             its contribution to stress sensitivity. Research in this
             area has both advanced basic science and generated broader
             lessons for studying complex diseases and traits. The
             authors evaluate four lines of evidence about the 5-HTT
             stress-sensitivity hypothesis: 1) observational studies
             about the serotonin transporter linked polymorphic region
             (5-HTTLPR), stress sensitivity, and depression in humans; 2)
             experimental neuroscience studies about the 5-HTTLPR and
             biological phenotypes relevant to the human stress response;
             3) studies of 5-HTT variation and stress sensitivity in
             nonhuman primates; and 4) studies of stress sensitivity and
             genetically engineered 5-HTT mutations in rodents. The
             authors then dispel some misconceptions and offer
             recommendations for GxE research. The authors discuss how
             GxE interaction hypotheses can be tested with large and
             small samples, how GxE research can be carried out before as
             well as after replicated gene discovery, the uses of GxE
             research as a tool for gene discovery, the importance of
             construct validation in evaluating GxE research, and the
             contribution of GxE research to the public understanding of
             genetic science.},
   Language = {eng},
   Doi = {10.1176/appi.ajp.2010.09101452},
   Key = {Caspi2010}
}

@article{Munoz2010,
   Author = {Muñoz, KE and Meyer-Lindenberg, A and Hariri, AR and Mervis, CB and Mattay, VS and Morris, CA and Berman, KF},
   Title = {Abnormalities in neural processing of emotional stimuli in
             Williams syndrome vary according to social vs. non-social
             content.},
   Journal = {NeuroImage},
   Volume = {50},
   Number = {1},
   Pages = {340-346},
   Address = {Section on Integrative Neuroimaging, National Institute of
             Mental Health, NIH, DHHS, Bethesda, MD 20892,
             USA.},
   Year = {2010},
   Month = {March},
   ISSN = {1053-8119},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20004252},
   Keywords = {Adult • Amygdala/physiopathology •
             Brain/*physiopathology • Brain Mapping •
             Cognition/*physiology • Emotions/*physiology •
             Female • Humans • Intelligence • Intelligence
             Tests • Magnetic Resonance Imaging • Male •
             Neural Pathways/physiopathology • Oxygen/blood •
             Prefrontal Cortex/physiopathology • *Social Behavior
             • Visual Perception/physiology • Williams
             Syndrome/*physiopathology},
   Abstract = {Williams syndrome (WS) is a rare genetic disorder caused by
             the deletion of approximately 25 genes on chromosome 7q11.23
             and is characterized by both hypersociability and increases
             in specific phobia and anticipatory anxiety regarding
             non-social entities or circumstances. Alterations in
             amygdala reactivity and prefrontal regulation consistent
             with the observed behavioral pattern of social versus
             non-social abnormalities have been previously demonstrated
             in individuals with WS (Meyer-Lindenberg et al., 2005).
             However, in that study, the social stimulus (faces) matching
             task was more difficult than the non-social scene (IAPS
             stimuli) matching task, making it impossible to disambiguate
             the relative contributions of task difficulty and stimulus
             type (social versus non-social). In the present study, we
             examined the performance of the same group of participants
             with WS and normal IQs during a more cognitively demanding
             task using the same scene stimuli as in the prior study.
             Confirming previous findings, the results indicated (a) a
             differential response of prefrontal regions as a function of
             task difficulty and (b) a persistently increased activation
             of the amygdala to non-social scenes by individuals with WS
             regardless of cognitive load. These data provide further
             evidence of disruption in amygdala-prefrontal circuitry in
             individuals with WS.},
   Language = {eng},
   Doi = {10.1016/j.neuroimage.2009.11.069},
   Key = {Munoz2010}
}

@article{Hariri2010,
   Author = {Hariri, AR},
   Title = {Genetic polymorphisms: a cornerstone of translational
             biobehavioral research.},
   Journal = {Science translational medicine},
   Volume = {2},
   Number = {18},
   Pages = {18ps6},
   Address = {Department of Psychology and Neuroscience, Institute for
             Genome Sciences and Policy, Duke University, Durham, NC
             27708, USA. ahmad.hariri@duke.edu},
   Year = {2010},
   Month = {February},
   ISSN = {1946-6234},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20371481},
   Keywords = {Amino Acid Substitution/genetics • Animals •
             *Behavior • Brain-Derived Neurotrophic Factor/genetics
             • Humans • Mice • *Polymorphism, Genetic
             • *Translational Medical Research},
   Abstract = {A new generation of interdisciplinary research seeks to use
             common functional genetic polymorphisms to model emergent
             variability in brain chemistry that regulates behaviorally
             relevant brain structure and function. This genetically
             mediated variability is then being mapped onto trajectories
             of risk for psychopathology, especially that precipitated by
             environmental adversity. This Perspective highlights a
             recent paper in Science that provides a powerful example of
             how a common functional genetic polymorphism can serve as a
             translational bridge between human and mouse research,
             extending our understanding of biological pathways that
             mediate individual differences in behavior and in risk for
             psychopathology.},
   Language = {eng},
   Doi = {10.1126/scitranslmed.3000811},
   Key = {Hariri2010}
}

@article{Manuck2010,
   Author = {Manuck, SB and Marsland, AL and Flory, JD and Gorka, A and Ferrell, RE and Hariri, AR},
   Title = {Salivary testosterone and a trinucleotide (CAG) length
             polymorphism in the androgen receptor gene predict amygdala
             reactivity in men.},
   Journal = {Psychoneuroendocrinology},
   Volume = {35},
   Number = {1},
   Pages = {94-104},
   Address = {Department of Psychology, University of Pittsburgh,
             Pittsburgh, PA 15260, United States. Manuck@pitt.edu},
   Year = {2010},
   Month = {January},
   ISSN = {0306-4530},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19493626},
   Keywords = {Adult • Amygdala/*physiology • DNA/genetics •
             Facial Expression • Genotype • Humans • Image
             Processing, Computer-Assisted • Individuality •
             Magnetic Resonance Imaging • Male • Middle Aged
             • Oxygen/blood • Polymorphism, Genetic •
             Receptors, Androgen/*genetics/physiology •
             Saliva/*metabolism • Social Perception •
             Testosterone/*metabolism • Trinucleotide
             Repeats/*genetics},
   Abstract = {In studies employing functional magnetic resonance imaging
             (fMRI), reactivity of the amygdala to threat-related sensory
             cues (viz., facial displays of negative emotion) has been
             found to correlate positively with interindividual
             variability in testosterone levels of women and young men
             and to increase on acute administration of exogenous
             testosterone. Many of the biological actions of testosterone
             are mediated by intracellular androgen receptors (ARs),
             which exert transcriptional control of androgen-dependent
             genes and are expressed in various regions of the brain,
             including the amygdala. Transactivation potential of the AR
             decreases (yielding relative androgen insensitivity) with
             expansion a polyglutamine stretch in the N-terminal domain
             of the AR protein, as encoded by a trinucleotide (CAG)
             repeat polymorphism in exon 1 of the X-chromosome AR gene.
             Here we examined whether amygdala reactivity to
             threat-related facial expressions (fear, anger) differs as a
             function of AR CAG length variation and endogenous
             (salivary) testosterone in a mid-life sample of 41 healthy
             men (mean age=45.6 years, range: 34-54 years; CAG repeats,
             range: 19-29). Testosterone correlated inversely with
             participant age (r=-0.39, p=0.012) and positively with
             number of CAG repeats (r=0.45, p=0.003). In partial
             correlations adjusted for testosterone level, reactivity in
             the ventral amygdala was lowest among men with largest
             number of CAG repeats. This inverse association was seen in
             both the right (r(p)=-0.34, p<0.05) and left (r(p)=-0.32,
             p<0.05) hemisphere. Activation of dorsal amygdala,
             correlated positively with individual differences in
             salivary testosterone, also in right (r=0.40, p<0.02) and
             left (r=0.32, p<0.05) hemisphere, but was not affected by
             number of CAG repeats. Hence, androgenic influences on
             threat-related reactivity in the ventral amygdala may be
             moderated partially by CAG length variation in the AR gene.
             Because individual differences in salivary testosterone also
             predicted dorsal amygdala reactivity and did so
             independently of CAG repeats, it is suggested that
             androgenic influences within this anatomically distinct
             region may be mediated, in part, by non-genomic or
             AR-independent mechanisms.},
   Language = {eng},
   Doi = {10.1016/j.psyneuen.2009.04.013},
   Key = {Manuck2010}
}

@article{Salgado-Pineda2010,
   Author = {Salgado-Pineda, P and Fakra, E and Delaveau, P and Hariri, AR and Blin,
             O},
   Title = {Differential patterns of initial and sustained responses in
             amygdala and cortical regions to emotional stimuli in
             schizophrenia patients and healthy participants.},
   Journal = {Journal of psychiatry & neuroscience : JPN},
   Volume = {35},
   Number = {1},
   Pages = {41-48},
   Address = {Centre d'Investigation Clinique-Unite de Pharmacologie
             Clinique et d'Evaluations Therapeutiques, Hopital de la
             Timone, Unite Mixte de Recherche, Centre national de la
             recherche scientifique, 6193 Institut de Neurosciences
             Cognitives de la Mediterranee, Ma},
   Year = {2010},
   Month = {January},
   ISSN = {1180-4882},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/20040245},
   Keywords = {Adult • Amygdala/*physiopathology • Brain Mapping
             • Cerebral Cortex/*physiopathology •
             Emotions/*physiology • Executive Function/*physiology
             • Facial Expression • Female • Humans •
             Magnetic Resonance Imaging • Male • Middle Aged
             • Neural Pathways/physiopathology •
             Neuropsychological Tests • Pattern Recognition,
             Visual/physiology • Psychiatric Status Rating Scales
             • Reaction Time • Schizophrenia/*physiopathology
             • Time Factors • Young Adult},
   Abstract = {<h4>Background</h4>We sought to investigate the altered
             brain responses to emotional stimuli in patients with
             schizophrenia.<h4>Methods</h4>We analyzed data from 14
             patients with schizophrenia and 14 healthy controls who
             performed an emotional face matching task. We evaluated
             brain activity and connectivity in the amygdala and cortical
             regions during the initial (first 21 seconds of each
             stimulation block) and sustained (last 21 seconds) stages of
             an emotional processing task, and we determined changes in
             amygdala activity across the emotional processing
             task.<h4>Results</h4>The patients with schizophrenia showed
             similar amygdala activation to the controls during the
             initial stage of processing, but their activation decreased
             during the sustained stage. The controls showed increasing
             amygdala activity across the emotional blocks, whereas
             activity progressively decreased in the schizophrenia group.
             The patients with schizophrenia showed increased cortical
             activity and interconnectivity in the medial frontal and
             inferior parietal cortex in the initial stage of emotional
             processing.There was increased activity in the superior
             temporal cortex and greater connectivity with the inferior
             parietal cortex in the sustained stage. Performance accuracy
             was lower in the schizophrenia group in the first part of
             the block, while their reaction time was longer in the
             latter part of the block.<h4>Limitations</h4>It was not
             possible to specify the moment at which the switch in
             amygdala response occurred.<h4>Conclusion</h4>Our findings
             suggest that patients with schizophrenia have an initial
             automatic emotional response but that they need to switch to
             a compensatory cognitive strategy to solve the
             task.},
   Language = {eng},
   Doi = {10.1503/jpn.090017},
   Key = {Salgado-Pineda2010}
}

@article{fds251972,
   Author = {Hariri, A},
   Title = {Imaging Genetics: Integration of Neuroimaging and Genetics
             in the Search for Predictive Markers},
   Pages = {532-537},
   Publisher = {Elsevier},
   Year = {2009},
   Month = {December},
   Doi = {10.1016/B978-0-12-369420-1.00047-0},
   Key = {fds251972}
}

@article{fds252002,
   Author = {Hariri, AR},
   Title = {Ahmad R. Hariri: award for distinguished scientific early
             career contributions to psychology.},
   Journal = {The American psychologist},
   Volume = {64},
   Number = {8},
   Pages = {683-684},
   Year = {2009},
   Month = {November},
   ISSN = {0003-066X},
   Doi = {10.1037/a0016290},
   Key = {fds252002}
}

@article{Blasi2009,
   Author = {Blasi, G and Hariri, AR and Alce, G and Taurisano, P and Sambataro, F and Das, S and Bertolino, A and Weinberger, DR and Mattay,
             VS},
   Title = {Preferential amygdala reactivity to the negative assessment
             of neutral faces.},
   Journal = {Biological psychiatry},
   Volume = {66},
   Number = {9},
   Pages = {847-853},
   Address = {Psychiatric Neuroscience Group, Department of Neurological
             and Psychiatric Sciences, University of Bari, Bari,
             Italy.},
   Year = {2009},
   Month = {November},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19709644},
   Keywords = {Adult • Amygdala/*physiology • Brain Mapping
             • Decision Making/*physiology • Emotions •
             *Facial Expression • Female • Gyrus
             Cinguli/physiology • Humans • Magnetic Resonance
             Imaging • Male • Neural Pathways/physiology •
             Prefrontal Cortex/physiology • Prejudice •
             Reaction Time • Social Perception},
   Abstract = {<h4>Background</h4>Prior studies suggest that the amygdala
             shapes complex behavioral responses to socially ambiguous
             cues. We explored human amygdala function during explicit
             behavioral decision making about discrete emotional facial
             expressions that can represent socially unambiguous and
             ambiguous cues.<h4>Methods</h4>During functional magnetic
             resonance imaging, 43 healthy adults were required to make
             complex social decisions (i.e., approach or avoid) about
             either relatively unambiguous (i.e., angry, fearful, happy)
             or ambiguous (i.e., neutral) facial expressions. Amygdala
             activation during this task was compared with that elicited
             by simple, perceptual decisions (sex discrimination) about
             the identical facial stimuli.<h4>Results</h4>Angry and
             fearful expressions were more frequently judged as avoidable
             and happy expressions most often as approachable. Neutral
             expressions were equally judged as avoidable and
             approachable. Reaction times to neutral expressions were
             longer than those to angry, fearful, and happy expressions
             during social judgment only. Imaging data on stimuli judged
             to be avoided revealed a significant task by emotion
             interaction in the amygdala. Here, only neutral facial
             expressions elicited greater activity during social judgment
             than during sex discrimination. Furthermore, during social
             judgment only, neutral faces judged to be avoided were
             associated with greater amygdala activity relative to
             neutral faces that were judged as approachable. Moreover,
             functional coupling between the amygdala and both
             dorsolateral prefrontal (social judgment > sex
             discrimination) and cingulate (sex discrimination > social
             judgment) cortices was differentially modulated by task
             during processing of neutral faces.<h4>Conclusions</h4>Our
             results suggest that increased amygdala reactivity and
             differential functional coupling with prefrontal circuitries
             may shape complex decisions and behavioral responses to
             socially ambiguous cues.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2009.06.017},
   Key = {Blasi2009}
}

@article{Fisher2009,
   Author = {Fisher, PM and Meltzer, CC and Price, JC and Coleman, RL and Ziolko, SK and Becker, C and Moses-Kolko, EL and Berga, SL and Hariri,
             AR},
   Title = {Medial prefrontal cortex 5-HT(2A) density is correlated with
             amygdala reactivity, response habituation, and functional
             coupling.},
   Journal = {Cerebral cortex (New York, N.Y. : 1991)},
   Volume = {19},
   Number = {11},
   Pages = {2499-2507},
   Address = {Center for Neuroscience, University of Pittsburgh,
             Pittsburgh, PA 15213, USA.},
   Year = {2009},
   Month = {November},
   ISSN = {1047-3211},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19321655},
   Keywords = {Adult • Amygdala/*physiology •
             Emotions/*physiology • Female • Habituation,
             Psychophysiologic/*physiology • Humans • Male
             • Neural Inhibition/*physiology • Prefrontal
             Cortex/*physiology • Receptor, Serotonin,
             5-HT2A/*metabolism • Tissue Distribution},
   Abstract = {Feedback inhibition of the amygdala via medial prefrontal
             cortex (mPFC) is an important component in the regulation of
             complex emotional behaviors. The functional dynamics of this
             corticolimbic circuitry are, in part, modulated by serotonin
             (5-HT). Serotonin 2A (5-HT(2A)) receptors within the mPFC
             represent a potential molecular mechanism through which 5-HT
             can modulate this corticolimbic circuitry. We employed a
             multimodal neuroimaging strategy to explore the relationship
             between threat-related amygdala reactivity, assessed using
             blood oxygen level-dependent functional magnetic resonance
             imaging, and mPFC 5-HT(2A) density, assessed using
             [(18)F]altanserin positron emission tomography in 35 healthy
             adult volunteers. We observed a significant inverse
             relationship wherein greater mPFC 5-HT(2A) density was
             associated with reduced threat-related right amygdala
             reactivity. Remarkably, 25-37% of the variability in
             amygdala reactivity was explained by mPFC 5-HT(2A) density.
             We also observed a positive correlation between mPFC
             5-HT(2A) density and the magnitude of right amygdala
             habituation. Furthermore, functional coupling between the
             amygdala and mPFC was positively correlated with 5-HT(2A)
             density suggesting that effective integration of emotionally
             salient information within this corticolimbic circuitry may
             be modulated, at least in part, by mPFC 5-HT(2A).
             Collectively, our results indicate that mPFC 5-HT(2A) is
             strongly associated with threat-related amygdala reactivity
             as well as its temporal habituation and functional coupling
             with prefrontal regulatory regions.},
   Language = {eng},
   Doi = {10.1093/cercor/bhp022},
   Key = {Fisher2009}
}

@article{Mechelli2009,
   Author = {Mechelli, A and Tognin, S and McGuire, PK and Prata, D and Sartori, G and Fusar-Poli, P and De Brito and S and Hariri, AR and Viding,
             E},
   Title = {Genetic vulnerability to affective psychopathology in
             childhood: a combined voxel-based morphometry and functional
             magnetic resonance imaging study.},
   Journal = {Biological psychiatry},
   Volume = {66},
   Number = {3},
   Pages = {231-237},
   Address = {Department of Psychology, Institute of Psychiatry, PO Box
             67, Division of Psychological Medicine and Psychiatry,
             King's College London, 103 Denmark Hill, London SE5 8AF,
             United Kingdom. a.mechelli@iop.kcl.ac.uk},
   Year = {2009},
   Month = {August},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19278671},
   Keywords = {Affective Disorders, Psychotic/*genetics/*pathology •
             Brain/*blood supply/*pathology • Brain Mapping •
             Catechol O-Methyltransferase/*genetics • Child •
             Genotype • Humans • Image Processing,
             Computer-Assisted/methods • Longitudinal Studies •
             Magnetic Resonance Imaging/methods • Male •
             Methionine/genetics • Neuropsychological Tests •
             Oxygen/blood • Photic Stimulation • Polymorphism,
             Genetic/*drug effects • Reaction Time/physiology •
             Twin Studies as Topic • Valine/genetics},
   Abstract = {<h4>Background</h4>The majority of affective psychopathology
             is rooted early in life and first emerges during childhood
             and adolescence. However, little is known about how genetic
             vulnerability affects brain structure and function in
             childhood since the vast majority of studies published so
             far have been conducted on adult participants. The present
             investigation examined for the first time the effects of
             catechol-O-methyltransferase (COMT) valine (val) 158
             methionine (met) (val158met) polymorphism, which has been
             shown to moderate predisposition to negative mood and
             affective disorders, on brain structure and function in
             children.<h4>Methods</h4>Voxel-based morphometry and
             functional magnetic resonance imaging were used to measure
             gray matter volume and emotional reactivity in 50 children
             aged between 10 and 12 years. We tested the hypothesis that
             met158 allele affects structural brain development and
             confers heightened reactivity within the affective
             frontolimbic circuit in children.<h4>Results</h4>The met158
             allele was positively associated with gray matter volume in
             the left hippocampal head where genotype accounted for 59%
             of interindividual variance. In addition, the met158 allele
             was positively associated with neuronal responses to fearful
             relative to neutral facial expressions in the right
             parahippocampal gyrus where genotype accounted for 14% of
             the interindividual variance.<h4>Conclusions</h4>These
             results indicate that the met158 allele is associated with
             increased gray matter volume and heightened reactivity
             during emotional processing within the limbic system in
             children as young as 10 to 12 years of age. These findings
             are consistent with the notion that genetic factors affect
             brain function to moderate vulnerability to affective
             psychopathology from childhood.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2009.01.033},
   Key = {Mechelli2009}
}

@article{Hariri2009a,
   Author = {Hariri, AR and Gorka, A and Hyde, LW and Kimak, M and Halder, I and Ducci,
             F and Ferrell, RE and Goldman, D and Manuck, SB},
   Title = {Divergent effects of genetic variation in endocannabinoid
             signaling on human threat- and reward-related brain
             function.},
   Journal = {Biological psychiatry},
   Volume = {66},
   Number = {1},
   Pages = {9-16},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, Pennsylvania 15213, USA. haririar@upmc.edu},
   Year = {2009},
   Month = {July},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19103437},
   Keywords = {Adult • Amidohydrolases/*genetics • Analysis of
             Variance • Anxiety/*genetics • Brain/anatomy \&
             histology/blood supply/*physiology • Brain Mapping
             • Cannabinoid Receptor Modulators/genetics •
             Endocannabinoids • Female • Genetic
             Variation/*genetics • Genotype • Humans •
             Image Processing, Computer-Assisted/methods • Magnetic
             Resonance Imaging/methods • Male • Middle Aged
             • Oxygen/blood • Psychiatric Status Rating Scales
             • Regression Analysis • *Reward • *Signal
             Transduction/genetics},
   Abstract = {<h4>Background</h4>Fatty acid amide hydrolase (FAAH) is a
             key enzyme in regulating endocannabinoid (eCB) signaling. A
             common single nucleotide polymorphism (C385A) in the human
             FAAH gene has been associated with increased risk for
             addiction and obesity.<h4>Methods</h4>Using imaging genetics
             in 82 healthy adult volunteers, we examined the effects of
             FAAH C385A on threat- and reward-related human brain
             function.<h4>Results</h4>Carriers of FAAH 385A, associated
             with reduced enzyme and possibly increased eCB signaling,
             had decreased threat-related amygdala reactivity but
             increased reward-related ventral striatal reactivity in
             comparison with C385 homozygotes. Similarly divergent
             effects of FAAH C385A genotype were manifest at the level of
             brain-behavior relationships. The 385A carriers showed
             decreased correlation between amygdala reactivity and trait
             anxiety but increased correlation between ventral striatal
             reactivity and delay discounting, an index of
             impulsivity.<h4>Conclusions</h4>Our results parallel
             pharmacologic and genetic dissection of eCB signaling, are
             consistent with the psychotropic effects of
             Delta(9)-tetrahydrocannabinol, and highlight specific neural
             mechanisms through which variability in eCB signaling
             impacts complex behavioral processes related to risk for
             addiction and obesity.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2008.10.047},
   Key = {Hariri2009a}
}

@article{Gianaros2009,
   Author = {Gianaros, PJ and Hariri, AR and Sheu, LK and Muldoon, MF and Sutton-Tyrrell, K and Manuck, SB},
   Title = {Preclinical atherosclerosis covaries with individual
             differences in reactivity and functional connectivity of the
             amygdala.},
   Journal = {Biological psychiatry},
   Volume = {65},
   Number = {11},
   Pages = {943-950},
   Address = {Department of Psychiatry and Psychology, University of
             Pittsburgh, 3811 O'Hara Street, Pittsburgh, Pennsylvania
             15213, USA. gianarospj@upmc.edu},
   Year = {2009},
   Month = {June},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19013557},
   Keywords = {Adult • Amygdala/blood supply/*physiopathology •
             Atherosclerosis/epidemiology/*pathology • Brain Mapping
             • Carotid Arteries/pathology • Female •
             Humans • Image Processing, Computer-Assisted/methods
             • *Individuality • Magnetic Resonance
             Imaging/methods • Male • Middle Aged •
             Oxygen/blood • Risk Factors • Statistics as Topic
             • Tunica Media/*pathology • Ultrasonography},
   Abstract = {<h4>Background</h4>Cardiovascular disease (CVD) is a major
             source of medical comorbidity for patients with mood and
             anxiety disorders, and it remains the leading public health
             burden for the general population in industrialized nations.
             Indirect neurobiological evidence suggests that preclinical
             risk for atherosclerosis, the main contributor to CVD, may
             be conferred by interindividual variation in the
             functionality of the amygdala, a brain system jointly
             involved in processing behaviorally salient stimuli and
             regulating the cardiovascular system.<h4>Methods</h4>In a
             neuroimaging study of 36 middle-aged adults (18 women) who
             were screened for confounding clinical cardiovascular and
             psychiatric disorders, we examined the direct covariation
             between a marker of preclinical atherosclerosis, carotid
             artery intima-media thickness (IMT), and interindividual
             variation in amygdala reactivity and functional connectivity
             assessed during the processing of behaviorally salient
             stimuli (angry and fearful facial expressions).<h4>Results</h4>After
             accounting for traditional CVD risk factors, a thickening of
             carotid IMT across individuals covaried with greater
             amygdala reactivity and a more positive functional
             connectivity between the amygdala and perigenual anterior
             cingulate cortex, a corticolimbic area also implicated in
             behavioral salience processing and cardiovascular
             regulation.<h4>Conclusions</h4>Individual differences in
             amygdala reactivity and functional connectivity may reflect
             facets of a novel, systems-level neural phenotype conferring
             risk for atherosclerosis and CVD.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2008.10.007},
   Key = {Gianaros2009}
}

@article{fds252001,
   Author = {Zhou, Z and Zhu, G and Hariri, AR and Enoch, MA and Scott, D and Sinha, R and Virkkunen, M and Mash, DC and Lipsky, RH and Hu, XZ and Hodgkinson, CA and Xu, K and Buzas, B and Yuan, Q and Shen, PH and Ferrell, RE and Manuck, SB and Brown, SM and Hauger, RL and Stohler, CS and Zubieta, JK and Goldman,
             D},
   Title = {Zhou et al. reply},
   Journal = {Nature},
   Volume = {458},
   Number = {7238},
   Pages = {E7-E7},
   Publisher = {Springer Nature},
   Year = {2009},
   Month = {April},
   ISSN = {0028-0836},
   Doi = {10.1038/nature07928},
   Key = {fds252001}
}

@article{Munoz2009,
   Author = {Gerber, AJ and Peterson, BS and Muñoz, KE and Hyde, LW and Hariri,
             AR},
   Title = {Imaging genetics.},
   Journal = {Journal of the American Academy of Child and Adolescent
             Psychiatry},
   Volume = {48},
   Number = {4},
   Pages = {356-361},
   Address = {Department of Psychology, Center for the Neural Basis of
             Cognition, University of Pittsburgh, Pittsburgh, PA
             15213-2593, USA.},
   Year = {2009},
   Month = {April},
   ISSN = {0890-8567},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19318879},
   Keywords = {Amygdala/metabolism • Environment • Gene
             Expression/genetics • Genetic Predisposition to Disease
             • Heterozygote Detection/*instrumentation • Humans
             • Mental Disorders/*genetics/metabolism •
             Molecular Biology/*methods • Serotonin/genetics •
             Temperament},
   Language = {eng},
   Doi = {10.1097/chi.0b013e31819aad07},
   Key = {Munoz2009}
}

@article{Drabant2009,
   Author = {Drabant, EM and McRae, K and Manuck, SB and Hariri, AR and Gross,
             JJ},
   Title = {Individual differences in typical reappraisal use predict
             amygdala and prefrontal responses.},
   Journal = {Biological psychiatry},
   Volume = {65},
   Number = {5},
   Pages = {367-373},
   Address = {Department of Psychology, Stanford University, Stanford,
             California 94305, USA.},
   Year = {2009},
   Month = {March},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18930182},
   Keywords = {Adult • Amygdala/*physiology •
             Emotions/*physiology • Facial Expression • Female
             • Humans • *Individuality • Middle Aged
             • Prefrontal Cortex/*physiology},
   Abstract = {<h4>Background</h4>Participants who are instructed to use
             reappraisal to downregulate negative emotion show decreased
             amygdala responses and increased prefrontal responses.
             However, it is not known whether individual differences in
             the tendency to use reappraisal manifests in similar neural
             responses when individuals are spontaneously confronted with
             negative situations. Such spontaneous emotion regulation
             might play an important role in normal and pathological
             responses to the emotional challenges of everyday
             life.<h4>Methods</h4>Fifty-six healthy women completed a
             blood oxygenation-level dependent functional magnetic
             resonance imaging challenge paradigm involving the
             perceptual processing of emotionally negative facial
             expressions. Participants also completed measures of typical
             emotion regulation use, trait anxiety, and
             neuroticism.<h4>Results</h4>Greater use of reappraisal in
             everyday life was related to decreased amygdala activity and
             increased prefrontal and parietal activity during the
             processing of negative emotional facial expressions. These
             associations were not attributable to variation in trait
             anxiety, neuroticism, or the use of another common form of
             emotion regulation, namely suppression.<h4>Conclusions</h4>These
             findings suggest that, like instructed reappraisal,
             individual differences in reappraisal use are associated
             with decreased activation in ventral emotion generative
             regions and increased activation in prefrontal control
             regions in response to negative stimuli. Such individual
             differences in emotion regulation might predict successful
             coping with emotional challenges as well as the onset of
             affective disorders.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2008.09.007},
   Key = {Drabant2009}
}

@article{Rasetti2009,
   Author = {Rasetti, R and Mattay, VS and Wiedholz, LM and Kolachana, BS and Hariri,
             AR and Callicott, JH and Meyer-Lindenberg, A and Weinberger,
             DR},
   Title = {Evidence that altered amygdala activity in schizophrenia is
             related to clinical state and not genetic
             risk.},
   Journal = {The American journal of psychiatry},
   Volume = {166},
   Number = {2},
   Pages = {216-225},
   Address = {Genes, Cognition, and Psychosis Program, IRP, NIMH, NIH, Rm.
             4S-235, 10 Center Dr., Bethesda, MD 20892,
             USA.},
   Year = {2009},
   Month = {February},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19074979},
   Keywords = {Adult • Amygdala/*physiopathology • Anger •
             Cognition Disorders/diagnosis/*genetics/*physiopathology
             • Dominance, Cerebral/physiology • Facial
             Expression • Fear/physiology • Female •
             Genetic Predisposition to Disease/*genetics • Gyrus
             Cinguli/physiopathology • Humans • *Image
             Processing, Computer-Assisted • Imaging,
             Three-Dimensional • *Magnetic Resonance Imaging •
             Male • Memory, Short-Term • Nerve
             Net/physiopathology • *Neuropsychological Tests •
             Oxygen/*blood • Pattern Recognition, Visual/physiology
             • Phenotype • Schizophrenia/diagnosis/*genetics/*physiopathology},
   Abstract = {<h4>Objective</h4>Although amygdala dysfunction is reported
             in schizophrenia, it is unknown whether this deficit
             represents a heritable phenotype that is related to risk for
             schizophrenia or whether it is related to disease state. The
             purpose of the present study was to examine amygdala
             response to threatening faces among healthy siblings of
             schizophrenia patients in whom a subtler heritable deficit
             might be observed.<h4>Method</h4>Participants were 34
             schizophrenia patients, 29 unaffected siblings, and 20
             healthy comparison subjects. Blood-oxygen-level-dependent
             (BOLD) functional magnetic resonance imaging (fMRI) was
             conducted during an implicit facial information processing
             task. The N-back working memory task, which has been shown
             to elicit prefrontal cortex abnormalities in unaffected
             siblings of schizophrenia patients, was employed as a
             positive experimental control.<h4>Results</h4>Schizophrenia
             patients demonstrated a deficit in amygdala reactivity to
             negative face stimuli and an alteration, correlated with
             neuroleptic drug dosage, in the functional coupling between
             the amygdala and subgenual cingulate. In contrast,
             unaffected siblings showed a pattern that was not
             statistically different from that of healthy comparison
             subjects. During the N-back working memory task, both
             schizophrenia patients and their unaffected siblings
             demonstrated a pattern of inefficient prefrontal cortex
             engagement, which is consistent with earlier evidence that
             this pattern is related to genetic risk for
             schizophrenia.<h4>Conclusions</h4>These data suggest that
             the pathophysiological mechanism underlying the inability of
             individuals with schizophrenia to normally engage the
             amygdala in processing fearful and angry facial
             representations is more likely a phenomenon related to the
             disease state, specifically to treatment.},
   Language = {eng},
   Doi = {10.1176/appi.ajp.2008.08020261},
   Key = {Rasetti2009}
}

@article{fds251971,
   Author = {Hariri, AR and Weinberger, DR},
   Title = {Genetics of Human Anxiety and Its Disorders},
   Pages = {669-677},
   Publisher = {Elsevier},
   Year = {2009},
   Month = {January},
   Abstract = {Normal variation in human emotionality, in temperament and
             risk for affective disorders, is explained to a large degree
             by genetic variation. The sequencing of the human genome has
             made it possible to test the role of specific genes on
             measures of human emotionality, on risk for affective
             disorders, and on the brain systems that appear to mediate
             emotion-related phenotypes. A novel approach, called imaging
             genetics, has shown that several genes that impact on brain
             serotonergic signaling affect the development and function
             of circuits of the limbic system involved in emotional
             experience and behavior. © 2009 Published by
             null.},
   Doi = {10.1016/B978-008045046-9.00845-7},
   Key = {fds251971}
}

@article{Forbes2009b,
   Author = {Forbes, EE and Brown, SM and Kimak, M and Ferrell, RE and Manuck, SB and Hariri, AR},
   Title = {Genetic variation in components of dopamine
             neurotransmission impacts ventral striatal reactivity
             associated with impulsivity.},
   Journal = {Molecular psychiatry},
   Volume = {14},
   Number = {1},
   Pages = {60-70},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, PA 15213, USA.},
   Year = {2009},
   Month = {January},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17893706},
   Keywords = {Adult • Analysis of Variance • Basal Ganglia/blood
             supply/*physiopathology • Case-Control Studies •
             Catechol O-Methyltransferase/genetics •
             Dopamine/*genetics/metabolism • Dopamine Plasma
             Membrane Transport Proteins/genetics • Female •
             Gene Frequency • Genetic Variation/*genetics •
             Genotype • Humans • Image Processing,
             Computer-Assisted/methods • Impulsive
             Behavior/*genetics/*pathology • Magnetic Resonance
             Imaging/methods • Male • Middle Aged •
             Oxygen/blood • Receptors, Dopamine D3/genetics •
             Receptors, Dopamine D4/genetics • Reward •
             Synaptic Transmission/*genetics • Young
             Adult},
   Abstract = {Individual differences in traits such as impulsivity involve
             high reward sensitivity and are associated with risk for
             substance use disorders. The ventral striatum (VS) has been
             widely implicated in reward processing, and individual
             differences in its function are linked to these disorders.
             Dopamine (DA) plays a critical role in reward processing and
             is a potent neuromodulator of VS reactivity. Moreover,
             altered DA signaling has been associated with normal and
             pathological reward-related behaviors. Functional
             polymorphisms in DA-related genes represent an important
             source of variability in DA function that may subsequently
             impact VS reactivity and associated reward-related
             behaviors. Using an imaging genetics approach, we examined
             the modulatory effects of common, putatively functional
             DA-related polymorphisms on reward-related VS reactivity
             associated with self-reported impulsivity. Genetic variants
             associated with relatively increased striatal DA release
             (DRD2 -141C deletion) and availability (DAT1 9-repeat), as
             well as diminished inhibitory postsynaptic DA effects (DRD2
             -141C deletion and DRD4 7-repeat), predicted 9-12% of the
             interindividual variability in reward-related VS reactivity.
             In contrast, genetic variation directly affecting DA
             signaling only in the prefrontal cortex (COMT Val158Met) was
             not associated with variability in VS reactivity. Our
             results highlight an important role for genetic
             polymorphisms affecting striatal DA neurotransmission in
             mediating interindividual differences in reward-related VS
             reactivity. They further suggest that altered VS reactivity
             may represent a key neurobiological pathway through which
             these polymorphisms contribute to variability in behavioral
             impulsivity and related risk for substance use
             disorders.},
   Language = {eng},
   Doi = {10.1038/sj.mp.4002086},
   Key = {Forbes2009b}
}

@article{Forbes2009a,
   Author = {Forbes, EE and Hariri, AR and Martin, SL and Silk, JS and Moyles, DL and Fisher, PM and Brown, SM and Ryan, ND and Birmaher, B and Axelson, DA and Dahl, RE},
   Title = {Altered striatal activation predicting real-world positive
             affect in adolescent major depressive disorder.},
   Journal = {The American journal of psychiatry},
   Volume = {166},
   Number = {1},
   Pages = {64-73},
   Address = {University of Pittsburgh School of Medicine, Western
             Psychiatric Institute and Clinic, 3811 O'Hara St., Loeffler
             319, Pittsburgh, PA 15213, USA. forbese@upmc.edu},
   Year = {2009},
   Month = {January},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19047324},
   Keywords = {Adolescent • Affect/*physiology • Age Factors
             • Caudate Nucleus/physiopathology • Child •
             Computers, Handheld • Corpus Striatum/*physiopathology
             • Depressive Disorder, Major/diagnosis/*physiopathology/psychology
             • Dominance, Cerebral/physiology • Female •
             Humans • *Image Processing, Computer-Assisted •
             *Magnetic Resonance Imaging • Male • Motivation
             • Neurons/physiology • Oxygen/*blood •
             Prefrontal Cortex/physiopathology • Prognosis •
             Reward • *Social Environment},
   Abstract = {<h4>Objective</h4>Alterations in reward-related brain
             function and phenomenological aspects of positive affect are
             increasingly examined in the development of major depressive
             disorder. The authors tested differences in reward-related
             brain function in healthy and depressed adolescents, and the
             authors examined direct links between reward-related brain
             function and positive mood that occurred in real-world
             contexts.<h4>Method</h4>Fifteen adolescents with major
             depressive disorder and 28 adolescents with no history of
             psychiatric disorder, ages 8-17 years, completed a
             functional magnetic resonance imaging guessing task
             involving monetary reward. Participants also reported their
             subjective positive affect in natural environments during a
             4-day cell-phone-based ecological momentary
             assessment.<h4>Results</h4>Adolescents with major depressive
             disorder exhibited less striatal response than healthy
             comparison adolescents during reward anticipation and reward
             outcome, but more response in dorsolateral and medial
             prefrontal cortex. Diminished activation in a caudate region
             associated with this depression group difference was
             correlated with lower subjective positive affect in natural
             environments, particularly within the depressed
             group.<h4>Conclusions</h4>Results support models of altered
             reward processing and related positive affect in young
             people with major depressive disorder and indicate that
             depressed adolescents' brain response to monetary reward is
             related to their affective experience in natural
             environments. Additionally, these results suggest that
             reward-processing paradigms capture brain function relevant
             to real-world positive affect.},
   Language = {eng},
   Doi = {10.1176/appi.ajp.2008.07081336},
   Key = {Forbes2009a}
}

@article{Fakra2009,
   Author = {Fakra, E and Hyde, LW and Gorka, A and Fisher, PM and Muñoz, KE and Kimak,
             M and Halder, I and Ferrell, RE and Manuck, SB and Hariri,
             AR},
   Title = {Effects of HTR1A C(-1019)G on amygdala reactivity and trait
             anxiety.},
   Journal = {Archives of general psychiatry},
   Volume = {66},
   Number = {1},
   Pages = {33-40},
   Address = {Hopital de laTimone, ServiceHospitalo-Universitaire
             dePsychiatrie, Hopital SteMarguerite, Marseille,
             France.},
   Year = {2009},
   Month = {January},
   ISSN = {0003-990X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19124686},
   Keywords = {Adult • *Alleles • Amygdala/*physiopathology
             • Anger/physiology • Anxiety Disorders/*genetics/physiopathology
             • Arousal/*genetics/physiology •
             Attention/physiology • Autoreceptors/*genetics •
             Dominance, Cerebral/physiology • Facial Expression
             • Fear/physiology • Female • Genotype •
             Humans • *Image Processing, Computer-Assisted •
             *Magnetic Resonance Imaging • Male • Middle Aged
             • Oxygen/*blood • Pattern Recognition,
             Visual/physiology • Receptor, Serotonin,
             5-HT1A/*genetics • Signal Transduction/genetics},
   Abstract = {<h4>Context</h4>Serotonin 1A (5-hydroxytryptamine 1A
             [5-HT(1A)]) autoreceptors mediate negative feedback
             inhibition of serotonergic neurons and play a critical role
             in regulating serotonin signaling involved in shaping the
             functional response of major forebrain targets, such as the
             amygdala, supporting complex behavioral processes. A common
             functional variation (C[-1019]G) in the human 5-HT(1A) gene
             (HTR1A) represents 1 potential source of such
             interindividual variability. Both in vitro and in vivo,
             -1019G blocks transcriptional repression, leading to
             increased autoreceptor expression. Thus, -1019G may
             contribute to relatively decreased serotonin signaling at
             postsynaptic forebrain target sites via increased negative
             feedback.<h4>Objectives</h4>To evaluate the effects of HTR1A
             C(-1019)G on amygdala reactivity and to use path analyses to
             explore the impact of HTR1A-mediated variability in amygdala
             reactivity on individual differences in trait anxiety. We
             hypothesized that -1019G, which potentially results in
             decreased serotonin signaling, would be associated with
             relatively decreased amygdala reactivity and related trait
             anxiety.<h4>Design</h4>Imaging genetics in participants from
             an archival database.<h4>Participants</h4>Eighty-nine
             healthy adults.<h4>Results</h4>Consistent with prior
             findings, -1019G was associated with significantly decreased
             threat-related amygdala reactivity. Importantly, this effect
             was independent of that associated with another common
             functional polymorphism that affects serotonin signaling,
             5-HTTLPR. While there were no direct genotype effects on
             trait anxiety, HTR1A C(-1019)G indirectly predicted 9.2% of
             interindividual variability in trait anxiety through its
             effects on amygdala reactivity.<h4>Conclusions</h4>Our
             findings further implicate relatively increased serotonin
             signaling, associated with a genetic variation that mediates
             increased 5-HT(1A) autoreceptors, in driving amygdala
             reactivity and trait anxiety. Moreover, they provide
             empirical documentation of the basic premise that genetic
             variation indirectly affects emergent behavioral processes
             related to psychiatric disease risk by biasing the response
             of underlying neural circuitries.},
   Language = {eng},
   Doi = {10.1001/archpsyc.66.1.33},
   Key = {Fakra2009}
}

@article{Hariri2009b,
   Author = {Hariri, AR},
   Title = {The neurobiology of individual differences in complex
             behavioral traits.},
   Journal = {Annual review of neuroscience},
   Volume = {32},
   Pages = {225-247},
   Address = {Department of Psychology and Neuroscience, Institute for
             Genome Sciences and Policy, Duke University, Durham, North
             Carolina 27708, USA. ah154@duke.edu},
   Year = {2009},
   Month = {January},
   ISSN = {0147-006X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19400720},
   Keywords = {Animals • Behavior/*physiology • Biological
             Markers/analysis • Brain Mapping/*methods •
             Genetic Predisposition to Disease/genetics • Genetic
             Variation/physiology • Humans • Magnetic Resonance
             Imaging/*methods • Neuropsychology/*methods •
             Positron-Emission Tomography/*methods • *Quantitative
             Trait, Heritable},
   Abstract = {Neuroimaging, especially BOLD fMRI, has begun to identify
             how variability in brain function contributes to individual
             differences in complex behavioral traits. In parallel,
             pharmacological fMRI and multimodal PET/fMRI are identifying
             how variability in molecular signaling pathways influences
             individual differences in brain function. Against this
             background, functional genetic polymorphisms are being
             utilized to understand the origins of variability in
             signaling pathways as well as to model efficiently how such
             emergent variability impacts behaviorally relevant brain
             function. This article provides an overview of a research
             strategy seeking to integrate these complementary
             technologies and utilizes existing empirical data to
             illustrate its effectiveness in illuminating the
             neurobiology of individual differences in complex behavioral
             traits. The article also discusses how such efforts can
             contribute to the identification of predictive markers that
             interact with environmental factors to precipitate disease
             and to develop more effective and individually tailored
             treatment regimes.},
   Language = {eng},
   Doi = {10.1146/annurev.neuro.051508.135335},
   Key = {Hariri2009b}
}

@article{Kienast2008,
   Author = {Kienast, T and Hariri, AR and Schlagenhauf, F and Wrase, J and Sterzer,
             P and Buchholz, HG and Smolka, MN and Gründer, G and Cumming, P and Kumakura, Y and Bartenstein, P and Dolan, RJ and Heinz,
             A},
   Title = {Dopamine in amygdala gates limbic processing of aversive
             stimuli in humans.},
   Journal = {Nature neuroscience},
   Volume = {11},
   Number = {12},
   Pages = {1381-1382},
   Address = {Department of Psychiatry and Psychotherapy, Campus Charite
             Mitte, Charite-University Medicine Berlin, Chariteplatz 1,
             10117 Berlin, Germany.},
   Year = {2008},
   Month = {December},
   ISSN = {1097-6256},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18978778},
   Keywords = {Adult • *Affect • Amygdala/blood
             supply/*metabolism/radionuclide imaging • *Brain
             Mapping • Dihydroxyphenylalanine/analogs \&
             derivatives/pharmacokinetics • Dopamine/*metabolism
             • Fluorine Radioisotopes/pharmacokinetics • Gyrus
             Cinguli/blood supply/radionuclide imaging • Humans
             • Image Processing, Computer-Assisted • Limbic
             System/*metabolism/radionuclide imaging • Magnetic
             Resonance Imaging/methods • Male • Middle Aged
             • Oxygen/blood • Positron-Emission
             Tomography/methods • Psychophysics},
   Abstract = {Dopamine is released under stress and modulates processing
             of aversive stimuli. We found that dopamine storage capacity
             in human amygdala, measured with 6-[(18)F]fluoro-L-DOPA
             positron emission tomography, was positively correlated with
             functional magnetic resonance imaging blood oxygen
             level-dependent signal changes in amygdala and dorsal
             anterior cingulate cortex that were evoked by aversive
             stimuli. Furthermore, functional connectivity between these
             two regions was inversely related to trait anxiety. Our
             results suggest that individual dopamine storage capacity in
             amygdala subserves modulation of emotional processing in
             amygdala and dorsal cingulate, thereby contributing to
             individual differences in anxious temperament.},
   Language = {eng},
   Doi = {10.1038/nn.2222},
   Key = {Kienast2008}
}

@article{Zanardi2008,
   Author = {Zanardi, R and Barbini, B and Rossini, D and Bernasconi, A and Fregni,
             F and Padberg, F and Rossi, S and Wirz-Justice, A and Terman, M and Martiny, K and Bersani, G and Hariri, AR and Pezawas, L and Roiser, JP and Bertolino, A and Calabrese, G and Magri, L and Benedetti, F and Pontiggia, A and Malaguti, A and Smeraldi, E and Colombo,
             C},
   Title = {New perspectives on techniques for the clinical
             psychiatrist: Brain stimulation, chronobiology and
             psychiatric brain imaging.},
   Journal = {Psychiatry and clinical neurosciences},
   Volume = {62},
   Number = {6},
   Pages = {627-637},
   Address = {San Raffaele Hospital Department of Psychiatry, Vita-Salute
             University, Milan, Italy. zanardi.raffaella@hsr.it},
   Year = {2008},
   Month = {December},
   ISSN = {1323-1316},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/19067998},
   Keywords = {Brain/*pathology • Diagnostic Imaging • Humans
             • Magnetic Resonance Imaging • Magnetic Resonance
             Spectroscopy • Mental Disorders/pathology/physiopathology/*therapy
             • Mood Disorders/psychology • Periodicity •
             Psychiatry/*methods/*trends • Tomography,
             Emission-Computed, Single-Photon • *Transcranial
             Magnetic Stimulation},
   Abstract = {This review summarizes a scientific dialogue between
             representatives in non-pharmacological treatment options of
             affective disorders. Among the recently introduced somatic
             treatments for depression those with most evidenced efficacy
             will be discussed. The first part of this article presents
             current opinions about the clinical applications of
             transcranial magnetic stimulation in the treatment of
             depression. The second part explains the most relevant uses
             of chronobiology in mood disorders, while the last part
             deals with the main perspectives on brain imaging techniques
             in psychiatry. The aim was to bridge gaps between the
             research evidence and clinical decisions, and reach an
             agreement on several key points of chronobiological and
             brain stimulation techniques, as well as on relevant
             objectives for future research.},
   Language = {eng},
   Doi = {10.1111/j.1440-1819.2008.01863.x},
   Key = {Zanardi2008}
}

@article{Bigos2008,
   Author = {Bigos, KL and Pollock, BG and Aizenstein, HJ and Fisher, PM and Bies,
             RR and Hariri, AR},
   Title = {Acute 5-HT reuptake blockade potentiates human amygdala
             reactivity.},
   Journal = {Neuropsychopharmacology : official publication of the
             American College of Neuropsychopharmacology},
   Volume = {33},
   Number = {13},
   Pages = {3221-3225},
   Address = {Department of Pharmaceutical Sciences, University of
             Pittsburgh, PA, USA. bigosk@mail.nih.gov},
   Year = {2008},
   Month = {December},
   ISSN = {0893-133X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18463627},
   Keywords = {Adult • Akathisia, Drug-Induced/metabolism/physiopathology
             • Amygdala/*drug effects/*metabolism •
             Anxiety/chemically induced/metabolism/physiopathology •
             Brain Chemistry/*drug effects/*physiology •
             Citalopram/*pharmacology • Cross-Over Studies •
             Depressive Disorder/drug therapy/metabolism/physiopathology
             • Dose-Response Relationship, Drug • Double-Blind
             Method • Facial Expression • Humans • Male
             • Middle Aged • Neuropsychological Tests •
             Photic Stimulation • Presynaptic Terminals/drug
             effects/metabolism • Serotonin/*metabolism •
             Serotonin Uptake Inhibitors/pharmacology • Young
             Adult},
   Abstract = {Variability in serotonin (5-HT) function is associated with
             individual differences in normal mood and temperament, as
             well as psychiatric illnesses, all of which are influenced
             by amygdala function. This study evaluated the acute effects
             of 5-HT reuptake blockade on amygdala function using
             pharmacological functional MRI. Eight healthy men completed
             a double-blind balanced crossover study with the selective
             5-HT reuptake inhibitor, citalopram (20 mg infused over 30
             min), and normal saline. Amygdala reactivity in response to
             novel facial expressions was assessed on three successive
             scans, once before drug/placebo infusion, once early in the
             infusion, and once at the end of infusion. Acute citalopram
             administration resulted in concentration-dependent increases
             in human amygdala reactivity to salient stimuli. The current
             pattern of 5-HT-mediated amygdala reactivity may represent
             an important pathway through which SSRIs achieve an
             antidepressant effect. Intriguingly, our data may also
             reveal a mechanism contributing to clinical observations of
             extreme agitation, restlessness, and suicidal ideation in
             some individuals during acute SSRI treatment. Developing a
             comprehensive model of how 5-HT modulates human amygdala
             reactivity supporting behavioral and physiological arousal
             will be instrumental for our understanding of basic
             neurobehavioral processes, their dysfunction in psychiatric
             illnesses, and their contribution to mechanism of treatment
             response.},
   Language = {eng},
   Doi = {10.1038/npp.2008.52},
   Key = {Bigos2008}
}

@misc{fds366149,
   Author = {Hariri, AR},
   Title = {Imaging Genetics: Integration of Neuroimaging and Genetics
             in the Search for Predictive Markers},
   Pages = {532-537},
   Booktitle = {Genomic and Personalized Medicine: V1-2},
   Year = {2008},
   Month = {November},
   ISBN = {9780123694201},
   Doi = {10.1016/B978-0-12-369420-1.00047-0},
   Key = {fds366149}
}

@article{Ousdal2008,
   Author = {Ousdal, OT and Jensen, J and Server, A and Hariri, AR and Nakstad, PH and Andreassen, OA},
   Title = {The human amygdala is involved in general behavioral
             relevance detection: evidence from an event-related
             functional magnetic resonance imaging Go-NoGo
             task.},
   Journal = {Neuroscience},
   Volume = {156},
   Number = {3},
   Pages = {450-455},
   Address = {TOP Project, Psychosis Research Section, Division of
             Psychiatry, Building 49, Ulleval University Hospital, Oslo,
             Norway.},
   Year = {2008},
   Month = {October},
   ISSN = {0306-4522},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18775476},
   Keywords = {Adult • Amygdala/*blood supply/*physiology •
             Decision Making/*physiology • Female • Humans
             • Image Processing, Computer-Assisted • *Magnetic
             Resonance Imaging • Male • Neuropsychological
             Tests • Oxygen/blood • Pattern Recognition, Visual
             • Photic Stimulation/methods • Reaction
             Time/physiology • Signal Detection,
             Psychological/*physiology • Young Adult},
   Abstract = {The amygdala is classically regarded as a detector of
             potential threat and as a critical component of the neural
             circuitry mediating conditioned fear responses. However, it
             has been reported that the human amygdala responds to
             multiple expressions of emotions as well as emotionally
             neutral stimuli of a novel, uncertain or ambiguous nature.
             Thus, it has been proposed that the function of the amygdala
             may be of a more general art, i.e. as a detector of
             behaviorally relevant stimuli [Sander D, Grafman J, Zalla T
             (2003) The human amygdala: an evolved system for relevance
             detection. Rev Neurosci 14:303-316]. To investigate this
             putative function of the amygdala, we used event related
             functional magnetic resonance imaging (fMRI) and a modified
             Go-NoGo task composed of behaviorally relevant and
             irrelevant letter and number stimuli. Analyses revealed
             bilateral amygdala activation in response to letter stimuli
             that were behaviorally relevant as compared with letters
             with less behavioral relevance. Similar results were
             obtained for relatively infrequent NoGo relevant stimuli as
             compared with more frequent Go stimuli. Our findings support
             a role for the human amygdala in general detection of
             behaviorally relevant stimuli.},
   Language = {eng},
   Doi = {10.1016/j.neuroscience.2008.07.066},
   Key = {Ousdal2008}
}

@article{Marsland2008,
   Author = {Marsland, AL and Gianaros, PJ and Abramowitch, SM and Manuck, SB and Hariri, AR},
   Title = {Interleukin-6 covaries inversely with hippocampal grey
             matter volume in middle-aged adults.},
   Journal = {Biological psychiatry},
   Volume = {64},
   Number = {6},
   Pages = {484-490},
   Address = {Behavioral Immunology Laboratory, University of Pittsburgh,
             PA 15260, USA. marsland@pitt.edu},
   Year = {2008},
   Month = {September},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18514163},
   Keywords = {Adipose Tissue/metabolism • Adult • Cognition
             Disorders/diagnosis/epidemiology • Diagnostic and
             Statistical Manual of Mental Disorders • Female •
             Hippocampus/*anatomy \& histology/*metabolism • Humans
             • Hypertension/epidemiology/metabolism •
             Inflammation/epidemiology/metabolism •
             Interleukin-6/*metabolism • Magnetic Resonance Imaging
             • Male • Middle Aged • Nerve Net/metabolism
             • Registries • Severity of Illness Index •
             Wechsler Scales},
   Abstract = {<h4>Background</h4>Converging animal findings suggest that
             higher peripheral levels of inflammation are associated with
             activation of central inflammatory mechanisms that result in
             hippocampal neurodegeneration and related impairment of
             memory function. We have recently shown, consistent with
             animal findings, an inverse association between peripheral
             levels of interleukin-6 (IL-6), a relatively stable marker
             of systemic inflammation, and memory function in mid-life
             adults. In the current study, we extend this work to test
             whether systemic inflammation is associated with reduced
             grey matter volume of the hippocampus.<h4>Methods</h4>For
             this purpose, we used a computational structural
             neuroimaging method (optimized voxel-based morphometry) to
             evaluate the relationship between plasma IL-6 levels and
             hippocampal grey matter volume in a sample of 76 relatively
             healthy community volunteers ages 30-54.<h4>Results</h4>Peripheral
             levels of IL-6 covaried inversely with hippocampal grey
             matter volume, and this relationship persisted after
             accounting for several possible confounders, including age,
             gender, race, years of education, percent body fat, blood
             pressure, smoking, physical activity, hours of sleep,
             alcohol use, and total grey matter volume.<h4>Conclusions</h4>To
             our knowledge, this is the first report of a relationship
             between a peripheral marker of IL-6 and hippocampal grey
             matter volume, raising the possibility that low-grade
             systemic inflammation could plausibly presage subclinical
             cognitive decline in part via structural neural
             pathways.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2008.04.016},
   Key = {Marsland2008}
}

@article{fds252000,
   Author = {Pezawas, L and Meyer-Lindenberg, A and Goldman, AL and Verchinski,
             BA and Chen, G and Kolachana, BS and Egan, MF and Mattay, VS and Hariri,
             AR and Weinberger, DR},
   Title = {MET BDNF protects against morphological S allele effects of
             5-HTTLPR},
   Journal = {Molecular Psychiatry},
   Volume = {13},
   Number = {7},
   Pages = {654},
   Publisher = {Springer Nature},
   Year = {2008},
   Month = {July},
   ISSN = {1359-4184},
   Doi = {10.1038/mp.2008.61},
   Key = {fds252000}
}

@article{Pezawas2008,
   Author = {Pezawas, L and Meyer-Lindenberg, A and Goldman, AL and Verchinski,
             BA and Chen, G and Kolachana, BS and Egan, MF and Mattay, VS and Hariri,
             AR and Weinberger, DR},
   Title = {Evidence of biologic epistasis between BDNF and SLC6A4 and
             implications for depression.},
   Journal = {Molecular psychiatry},
   Volume = {13},
   Number = {7},
   Pages = {709-716},
   Address = {Genes, Cognition and Psychosis Program, National Institute
             of Mental Health, National Institutes of Health, Bethesda,
             MD 20892, USA.},
   Year = {2008},
   Month = {July},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18347599},
   Keywords = {Amino Acid Substitution • Brain/pathology •
             Brain-Derived Neurotrophic Factor/*genetics •
             Depression/*genetics/pathology • Depressive
             Disorder/*genetics/pathology • *Epistasis, Genetic
             • European Continental Ancestry Group/genetics •
             Gyrus Cinguli/pathology • Humans • Magnetic
             Resonance Imaging • Polymorphism, Genetic •
             Reference Values • Serotonin Plasma Membrane Transport
             Proteins/*genetics},
   Abstract = {Complex genetic disorders such as depression likely exhibit
             epistasis, but neural mechanisms of such gene-gene
             interactions are incompletely understood. 5-HTTLPR and BDNF
             VAL66MET, functional polymorphisms of the serotonin (5-HT)
             transporter (SLC6A4) and brain-derived neurotrophic factor
             (BDNF) gene, impact on two distinct, but interacting
             signaling systems, which have been related to depression and
             to the modulation of neurogenesis and plasticity of
             circuitries of emotion processing. Recent clinical studies
             suggest that the BDNF MET allele, which shows abnormal
             intracellular trafficking and regulated secretion, has a
             protective effect regarding the development of depression
             and in mice of social defeat stress. Here we show, using
             anatomical neuroimaging techniques in a sample of healthy
             subjects (n=111), that the BDNF MET allele, which is
             predicted to have reduced responsivity to 5-HT signaling,
             protects against 5-HTTLPR S allele-induced effects on a
             brain circuitry encompassing the amygdala and the subgenual
             portion of the anterior cingulate (rAC). Our analyses
             revealed no effect of the 5-HTTLPR S allele on rAC volume in
             the presence of BDNF MET alleles, whereas a significant
             volume reduction (P<0.001) was seen on BDNF VAL/VAL
             background. Interacting genotype effects were also found in
             structural connectivity between amygdala and rAC (P=0.002).
             These data provide in vivo evidence of biologic epistasis
             between SLC6A4 and BDNF in the human brain by identifying a
             neural mechanism linking serotonergic and neurotrophic
             signaling on the neural systems level, and have implications
             for personalized treatment planning in depression.},
   Language = {eng},
   Doi = {10.1038/mp.2008.32},
   Key = {Pezawas2008}
}

@article{Gianaros2008b,
   Author = {Gianaros, PJ and Horenstein, JA and Hariri, AR and Sheu, LK and Manuck,
             SB and Matthews, KA and Cohen, S},
   Title = {Potential neural embedding of parental social
             standing.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {3},
   Number = {2},
   Pages = {91-96},
   Address = {Department of Psychiatry, University of Pittsburgh, 3811
             O'Hara Street, Pittsburgh, PA 15213, USA.
             gianarospj@upmc.edu},
   Year = {2008},
   Month = {June},
   ISSN = {1749-5016},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18594696},
   Keywords = {Adaptation, Psychological/physiology •
             Aggression/*physiology/psychology •
             Amygdala/*physiology • Facial Expression •
             Hierarchy, Social • Humans • Magnetic Resonance
             Imaging • Male • Parents/psychology •
             Reference Values • Self Concept • *Social Class
             • *Social Perception • Stress,
             Psychological/*physiopathology • Young
             Adult},
   Abstract = {Socioeconomic disadvantage during childhood and adolescence
             predicts poor mental and physical health and premature death
             by major medical diseases in adulthood. However, the neural
             pathways through which socioeconomic factors may exert a
             developmental influence on health and longevity remain
             largely unknown. This fMRI study provides novel evidence of
             a unique relationship between the perception that one's
             parents had a relatively low social standing--a putative
             indicator of early socioeconomic disadvantage--and greater
             amygdala reactivity to threatening facial expressions. This
             relationship was not explained by several possible
             confounders, including sex, ethnicity, dispositional
             emotionality, symptoms of depression and anxiety, parental
             education and participants' perceptions of their own social
             standing. The amygdala expresses marked developmental
             plasticity and plays instrumental roles in processing
             emotional information, regulating emotion-related behaviors
             and orchestrating biobehavioral stress responses throughout
             life. Thus, these findings may provide insight into the
             neurodevelopmental pathways impacting socioeconomic
             disparities in health.},
   Language = {eng},
   Doi = {10.1093/scan/nsn003},
   Key = {Gianaros2008b}
}

@article{Fisher2008,
   Author = {Fisher, PM and Muñoz, KE and Hariri, AR},
   Title = {Identification of neurogenetic pathways of risk for
             psychopathology.},
   Journal = {American journal of medical genetics. Part C, Seminars in
             medical genetics},
   Volume = {148C},
   Number = {2},
   Pages = {147-153},
   Address = {University of Pittsburgh, Pittsburgh, PA 15213-2593,
             USA.},
   Year = {2008},
   Month = {May},
   ISSN = {1552-4868},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18412103},
   Keywords = {Brain/growth \& development/*physiopathology • Catechol
             O-Methyltransferase/genetics/physiology • Diagnostic
             Imaging • Dopamine Plasma Membrane Transport
             Proteins/genetics/physiology • Female • Humans
             • Male • Mental Disorders/diagnosis/*genetics/*physiopathology
             • Monoamine Oxidase/genetics/physiology •
             Psychopathology • Risk Factors • Serotonin Plasma
             Membrane Transport Proteins/genetics/physiology •
             Tryptophan Hydroxylase/genetics/physiology},
   Abstract = {Imaging genetics has been a highly effective and
             increasingly applied strategy for identifying the impact of
             genetic polymorphisms on individual differences in neural
             circuitry supporting complex behaviors. The application of
             imaging genetics towards further elucidating neural
             circuitry associated with the pathophysiology of psychiatric
             illness is of particular interest given its potential to
             guide the development and improvement of current therapeutic
             methods. The identification of genetic variants that
             contribute to or predict the disruption of specific neural
             pathways associated with psychopathology may also serve as
             useful markers of risk demarcating individuals with elevated
             susceptibility for psychiatric illness and affording early
             or even preemptive treatment strategies. In the continued
             development of this technique, recent multimodal
             neuroimaging strategies and studies examining the effects of
             multiple genes in concert within large subject populations
             have shown promise in the development of a more complete
             understanding of the interrelationships between genes, brain
             function, behavior and associated risk for
             psychopathology.},
   Language = {eng},
   Doi = {10.1002/ajmg.c.30173},
   Key = {Fisher2008}
}

@article{Munafo2008,
   Author = {Munafò, MR and Brown, SM and Hariri, AR},
   Title = {Serotonin transporter (5-HTTLPR) genotype and amygdala
             activation: a meta-analysis.},
   Journal = {Biological psychiatry},
   Volume = {63},
   Number = {9},
   Pages = {852-857},
   Address = {Department of Experimental Psychology, University of
             Bristol, United Kingdom. marcus.munafo@bristol.ac.uk},
   Year = {2008},
   Month = {May},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17949693},
   Keywords = {Adolescent • Adult • Alleles •
             Amygdala/*physiopathology • Anxiety
             Disorders/genetics/physiopathology • Depressive
             Disorder/genetics/physiopathology • Female •
             *Genotype • Humans • Individuality • Magnetic
             Resonance Imaging • Male • Phenotype •
             Polymorphism, Genetic/*genetics • Promoter Regions,
             Genetic/*genetics • Serotonin Plasma Membrane Transport
             Proteins/*genetics • Stress, Psychological/complications},
   Abstract = {<h4>Background</h4>We evaluated the magnitude of the
             reported associations between amygdala activation and the
             serotonin transporter gene linked polymorphic region
             (5-HTTLPR) and the likely effect size of this
             relationship.<h4>Methods</h4>We used meta-analytic
             techniques to combine data from existing published and
             unpublished studies. We also tested for possible publication
             bias and explored possible moderating influences on any
             association, such as sample ancestry.<h4>Results</h4>Our
             results provide support for the association of the 5-HTTLPR
             polymorphism and amygdala activation and suggest that this
             locus may account for up to 10% of phenotypic variance.
             Although we did not observe evidence for potential
             publication bias in our main analysis, this was due in part
             to efforts to obtain unpublished data pertinent to this
             meta-analysis, and when three unpublished data sets were
             excluded we did observe evidence of such bias. We also
             observed evidence that the first published study may provide
             an overestimate of the true effect size, which is consistent
             with findings from genetic association studies of other
             phenotypes.<h4>Conclusions</h4>Although our analysis
             provides support for the association of the 5-HTTLPR
             polymorphism and amygdala activation, it also suggests that
             most studies to date are nevertheless lacking in statistical
             power. Increasing the sample sizes of future imaging
             genetics studies will allow a more accurate characterization
             of any true effect size and afford adequate power to examine
             the impact of multiple polymorphisms that likely work in
             concert to affect gene function and, in turn, bias neural
             processes mediating dispositional traits such as temperament
             and personality.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2007.08.016},
   Key = {Munafo2008}
}

@article{Krystal2008,
   Author = {Krystal, JH and Carter, CS and Geschwind, D and Manji, HK and March, JS and Nestler, EJ and Zubieta, J-K and Charney, DS and Goldman, D and Gur, RE and Lieberman, JA and Roy-Byrne, P and Rubinow, DR and Anderson, SA and Barondes, S and Berman, KF and Blair, J and Braff, DL and Brown, ES and Calabrese, JR and Carlezon, WA and Cook, EH and Davidson, RJ and Davis,
             M and Desimone, R and Drevets, WC and Duman, RS and Essock, SM and Faraone,
             SV and Freedman, R and Friston, KJ and Gelernter, J and Geller, B and Gill,
             M and Gould, E and Grace, AA and Grillon, C and Gueorguieva, R and Hariri,
             AR and Innis, RB and Jones, EG and Kleinman, JE and Koob, GF and Krystal,
             AD and Leibenluft, E and Levinson, DF and Levitt, PR and Lewis, DA and Liberzon, I and Lipska, BK and Marder, SR and Markou, A and Mason, GF and McDougle, CJ and McEwen, BS and McMahon, FJ and Meaney, MJ and Meltzer,
             HY and Merikangas, KR and Meyer-Lindenberg, A and Mirnics, K and Monteggia, LM and Neumeister, A and O'Brien, CP and Owen, MJ and Pine,
             DS and Rapoport, JL and Rauch, SL and Robbins, TW and Rosenbaum, JF and Rosenberg, DR and Ross, CA and Rush, AJ and Sackeim, HA and Sanacora, G and Schatzberg, AF and Shaham, Y and Siever, LJ and Sunderland, T and Tecott, LH and Thase, ME and Todd, RD and Weissman, MM and Yehuda, R and Yoshikawa, T and Young, EA and McCandless, R},
   Title = {It is time to take a stand for medical research and against
             terrorism targeting medical scientists.},
   Journal = {Biol Psychiatry},
   Volume = {63},
   Number = {8},
   Pages = {725-727},
   Address = {Department of Psychiatry, Yale University School of
             Medicine, New Haven, Connecticut, USA. john.krystal@yale.edu},
   Year = {2008},
   Month = {April},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18371494},
   Keywords = {*Animal Experimentation • Animal Rights • Animals
             • *Attitude of Health Personnel • *Biomedical
             Research • Crime/prevention \& control • Ethics,
             Research • Humans • Primates • *Research
             Personnel • Terrorism/*prevention \& control •
             United States},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2008.03.005},
   Key = {Krystal2008}
}

@article{Zhou2008,
   Author = {Zhou, Z and Zhu, G and Hariri, AR and Enoch, M-A and Scott, D and Sinha, R and Virkkunen, M and Mash, DC and Lipsky, RH and Hu, X-Z and Hodgkinson, CA and Xu, K and Buzas, B and Yuan, Q and Shen, P-H and Ferrell, RE and Manuck,
             SB and Brown, SM and Hauger, RL and Stohler, CS and Zubieta, J-K and Goldman, D},
   Title = {Genetic variation in human NPY expression affects stress
             response and emotion.},
   Journal = {Nature},
   Volume = {452},
   Number = {7190},
   Pages = {997-1001},
   Address = {Laboratory of Neurogenetics, NIAAA, NIH, Bethesda, Maryland
             20892, USA.},
   Year = {2008},
   Month = {April},
   ISSN = {0028-0836},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18385673},
   Keywords = {Alleles • Anxiety/genetics • Anxiety
             Disorders/genetics • Brain/*metabolism/physiology/physiopathology
             • *Emotions • European Continental Ancestry
             Group/genetics • Facial Expression •
             Finland/ethnology • Gene Expression
             Regulation/*genetics • Genetic Variation/*genetics
             • Haplotypes/genetics • Humans •
             Lymphocytes/metabolism • Magnetic Resonance Imaging
             • Male • Neuropeptide Y/blood/*genetics •
             Opioid Peptides/metabolism • Pain/genetics •
             Polymorphism, Single Nucleotide/genetics • RNA,
             Messenger/genetics/metabolism • Stress,
             Physiological/*genetics/psychology • United
             States/ethnology},
   Abstract = {Understanding inter-individual differences in stress
             response requires the explanation of genetic influences at
             multiple phenotypic levels, including complex behaviours and
             the metabolic responses of brain regions to emotional
             stimuli. Neuropeptide Y (NPY) is anxiolytic and its release
             is induced by stress. NPY is abundantly expressed in regions
             of the limbic system that are implicated in arousal and in
             the assignment of emotional valences to stimuli and
             memories. Here we show that haplotype-driven NPY expression
             predicts brain responses to emotional and stress challenges
             and also inversely correlates with trait anxiety. NPY
             haplotypes predicted levels of NPY messenger RNA in
             post-mortem brain and lymphoblasts, and levels of plasma
             NPY. Lower haplotype-driven NPY expression predicted higher
             emotion-induced activation of the amygdala, as well as
             diminished resiliency as assessed by pain/stress-induced
             activations of endogenous opioid neurotransmission in
             various brain regions. A single nucleotide polymorphism (SNP
             rs16147) located in the promoter region alters NPY
             expression in vitro and seems to account for more than half
             of the variation in expression in vivo. These convergent
             findings are consistent with the function of NPY as an
             anxiolytic peptide and help to explain inter-individual
             variation in resiliency to stress, a risk factor for many
             diseases.},
   Language = {eng},
   Doi = {10.1038/nature06858},
   Key = {Zhou2008}
}

@article{Fakra2008,
   Author = {Fakra, E and Salgado-Pineda, P and Delaveau, P and Hariri, AR and Blin,
             O},
   Title = {Neural bases of different cognitive strategies for facial
             affect processing in schizophrenia.},
   Journal = {Schizophrenia research},
   Volume = {100},
   Number = {1-3},
   Pages = {191-205},
   Address = {CIC-UPCET, Hopital de la Timone, UMR CNRS 6193 INCM,
             Marseille, France. eric.fakra@ap-hm.fr},
   Year = {2008},
   Month = {March},
   ISSN = {0920-9964},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18234477},
   Keywords = {Adult • Affect/*physiology • Amygdala/physiopathology
             • Brain/*physiopathology • Brain Mapping •
             Cerebral Cortex/physiopathology • Control Groups •
             *Facial Expression • Female • Functional
             Laterality/physiology • Humans • Limbic
             System/physiopathology • Magnetic Resonance
             Imaging/*statistics \& numerical data • Male •
             Models, Neurological • Neural Pathways/physiopathology
             • Recognition (Psychology)/physiology •
             Schizophrenia/*diagnosis/physiopathology •
             *Schizophrenic Psychology • Social Perception •
             Task Performance and Analysis • Visual
             Perception/*physiology},
   Abstract = {<h4>Objective</h4>To examine the neural basis and dynamics
             of facial affect processing in schizophrenic patients as
             compared to healthy controls.<h4>Method</h4>Fourteen
             schizophrenic patients and fourteen matched controls
             performed a facial affect identification task during fMRI
             acquisition. The emotional task included an intuitive
             emotional condition (matching emotional faces) and a more
             cognitively demanding condition (labeling emotional faces).
             Individual analysis for each emotional condition, and
             second-level t-tests examining both within-, and
             between-group differences, were carried out using a random
             effects approach. Psychophysiological interactions (PPI)
             were tested for variations in functional connectivity
             between amygdala and other brain regions as a function of
             changes in experimental conditions (labeling versus
             matching).<h4>Results</h4>During the labeling condition,
             both groups engaged similar networks. During the matching
             condition, schizophrenics failed to activate regions of the
             limbic system implicated in the automatic processing of
             emotions. PPI revealed an inverse functional connectivity
             between prefrontal regions and the left amygdala in healthy
             volunteers but there was no such change in patients.
             Furthermore, during the matching condition, and compared to
             controls, patients showed decreased activation of regions
             involved in holistic face processing (fusiform gyrus) and
             increased activation of regions associated with feature
             analysis (inferior parietal cortex, left middle temporal
             lobe, right precuneus).<h4>Conclusions</h4>Our findings
             suggest that schizophrenic patients invariably adopt a
             cognitive approach when identifying facial affect. The
             distributed neocortical network observed during the
             intuitive condition indicates that patients may resort to
             feature-based, rather than configuration-based, processing
             and may constitute a compensatory strategy for limbic
             dysfunction.},
   Language = {eng},
   Doi = {10.1016/j.schres.2007.11.040},
   Key = {Fakra2008}
}

@article{Buckholtz2008,
   Author = {Buckholtz, JW and Callicott, JH and Kolachana, B and Hariri, AR and Goldberg, TE and Genderson, M and Egan, MF and Mattay, VS and Weinberger, DR and Meyer-Lindenberg, A},
   Title = {Genetic variation in MAOA modulates ventromedial prefrontal
             circuitry mediating individual differences in human
             personality.},
   Journal = {Molecular psychiatry},
   Volume = {13},
   Number = {3},
   Pages = {313-324},
   Address = {Neuroimaging Core Facility, National Institute for Mental
             Health, NIH, DHHS, Bethesda, MD 20892-1365,
             USA.},
   Year = {2008},
   Month = {March},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17519928},
   Keywords = {Adult • Brain Mapping • Facial Expression •
             Female • *Genetic Variation • Humans • Image
             Processing, Computer-Assisted • *Individuality •
             Magnetic Resonance Imaging • Male • Models,
             Biological • Monoamine Oxidase/*genetics • Neural
             Pathways/blood supply/physiology • Neuropsychological
             Tests • Oxygen/blood • Personality/*genetics
             • Photic Stimulation/methods • Prefrontal
             Cortex/blood supply/*physiology},
   Abstract = {Little is known about neural mechanisms underlying human
             personality and temperament, despite their considerable
             importance as highly heritable risk mediators for somatic
             and psychiatric disorders. To identify these circuits, we
             used a combined genetic and imaging approach focused on
             Monoamine Oxidase A (MAOA), encoding a key enzyme for
             monoamine metabolism previously associated with temperament
             and antisocial behavior. Male carriers of a low-expressing
             genetic variant exhibited dysregulated amygdala activation
             and increased functional coupling with ventromedial
             prefrontal cortex (vmPFC). Stronger coupling predicted
             increased harm avoidance and decreased reward dependence
             scores, suggesting that this circuitry mediates a part of
             the association of MAOA with these traits. We utilized path
             analysis to parse the effective connectivity within this
             system, and provide evidence that vmPFC regulates amygdala
             indirectly by influencing rostral cingulate cortex function.
             Our data implicate a neural circuit for variation in human
             personality under genetic control, provide an anatomically
             consistent mechanism for vmPFC-amygdala interactions
             underlying this variation, and suggest a role for vmPFC as a
             superordinate regulatory area for emotional arousal and
             social behavior.},
   Language = {eng},
   Doi = {10.1038/sj.mp.4002020},
   Key = {Buckholtz2008}
}

@article{fds251999,
   Author = {Hariri, AR},
   Title = {Imaging genetics offers new predictive markers of individual
             differences in behavior and risk for psychiatric
             diseases},
   Journal = {Neuropsychopharmacology},
   Volume = {33},
   Number = {1},
   Pages = {201-202},
   Publisher = {Springer Nature},
   Year = {2008},
   Month = {January},
   ISSN = {0893-133X},
   Doi = {10.1038/sj.npp.1301608},
   Key = {fds251999}
}

@article{Gianaros2008a,
   Author = {Gianaros, PJ and Sheu, LK and Matthews, KA and Jennings, JR and Manuck,
             SB and Hariri, AR},
   Title = {Individual differences in stressor-evoked blood pressure
             reactivity vary with activation, volume, and functional
             connectivity of the amygdala.},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {28},
   Number = {4},
   Pages = {990-999},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, Pennsylvania 15213, USA. gianarospj@upmc.edu},
   Year = {2008},
   Month = {January},
   ISSN = {0270-6474},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18216206},
   Keywords = {Adolescent • Adult • Amygdala/anatomy \&
             histology/*physiology • Blood Pressure/*physiology
             • Female • Humans • *Individuality •
             Male • Nerve Net/anatomy \& histology/*physiology
             • Organ Size/physiology • Pons/anatomy \&
             histology/physiology • Stress, Psychological/*physiopathology},
   Abstract = {Individuals who exhibit exaggerated blood pressure reactions
             to psychological stressors are at risk for hypertension,
             ventricular hypertrophy, and premature atherosclerosis;
             however, the neural systems mediating exaggerated blood
             pressure reactivity and associated cardiovascular risk in
             humans remain poorly defined. Animal models indicate that
             the amygdala orchestrates stressor-evoked blood pressure
             reactions via reciprocal signaling with corticolimbic and
             brainstem cardiovascular-regulatory circuits. Based on these
             models, we used a multimodal neuroimaging approach to
             determine whether human individual differences in
             stressor-evoked blood pressure reactivity vary with amygdala
             activation, gray matter volume, and functional connectivity
             with corticolimbic and brainstem areas implicated in
             stressor processing and cardiovascular regulation. We
             monitored mean arterial pressure (MAP) and concurrent
             functional magnetic resonance imaging BOLD signal changes in
             healthy young individuals while they completed a Stroop
             color-word stressor task, validated previously in
             epidemiological studies of cardiovascular risk. Individuals
             exhibiting greater stressor-evoked MAP reactivity showed (1)
             greater amygdala activation, (2) lower amygdala gray matter
             volume, and (3) stronger positive functional connectivity
             between the amygdala and perigenual anterior cingulate
             cortex and brainstem pons. Individual differences in
             amygdala activation, gray matter volume, and functional
             connectivity with corticolimbic and brainstem circuits may
             partly underpin cardiovascular disease risk by impacting
             stressor-evoked blood pressure reactivity.},
   Language = {eng},
   Doi = {10.1523/jneurosci.3606-07.2008},
   Key = {Gianaros2008a}
}

@article{Bigos2007,
   Author = {Bigos, KL and Hariri, AR},
   Title = {Neuroimaging: technologies at the interface of genes, brain,
             and behavior.},
   Journal = {Neuroimaging clinics of North America},
   Volume = {17},
   Number = {4},
   Pages = {459-viii},
   Address = {Department of Pharmaceutical Sciences, University of
             Pittsburgh School of Pharmacy, Pittsburgh, PA 15213,
             USA.},
   Year = {2007},
   Month = {November},
   ISSN = {1052-5149},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17983963},
   Keywords = {Behavior/*physiology • Brain/*physiology • *Brain
             Mapping • *Gene Expression • Humans •
             *Polymorphism, Genetic},
   Abstract = {Neuroimaging technologies provide a powerful approach to
             exploring the genetic basis of individual differences in
             complex behaviors and vulnerability to neuropsychiatric
             illness. Functional MRI studies have established important
             physiologic links between genetic polymorphisms and robust
             differences in information processing within distinct brain
             regions and circuits that have been linked to the
             manifestation of various disease. Neuroimaging technologies
             represent a critical tool in efforts to understand the
             neurobiology of normal and pathologic behavioral states.
             Research capitalizing on neuroimagingbased integration will
             contribute to the identification of predictive markers and
             biologic pathways for neuropsychiatric disease vulnerability
             and the generation of novel targets for therapeutic
             intervention.},
   Language = {eng},
   Doi = {10.1016/j.nic.2007.09.005},
   Key = {Bigos2007}
}

@article{Manuck2007,
   Author = {Manuck, SB and Brown, SM and Forbes, EE and Hariri,
             AR},
   Title = {Temporal stability of individual differences in amygdala
             reactivity.},
   Journal = {The American journal of psychiatry},
   Volume = {164},
   Number = {10},
   Pages = {1613-1614},
   Year = {2007},
   Month = {October},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17898358},
   Keywords = {Adult • Amygdala/*physiology • Anger/physiology
             • Emotions/*physiology • Fear/physiology •
             Female • Humans • *Individuality • Magnetic
             Resonance Imaging/*statistics \& numerical data • Male
             • Middle Aged • Mood Disorders/diagnosis},
   Language = {eng},
   Doi = {10.1176/appi.ajp.2007.07040609},
   Key = {Manuck2007}
}

@article{fds251998,
   Author = {Rubino, V and Blasi, G and Latorre, V and Fazio, L and d'Errico, I and Mazzola, V and Caforio, G and Nardini, M and Popolizio, T and Hariri, A and Arciero, G and Bertolino, A},
   Title = {Activity in medial prefrontal cortex during cognitive
             evaluation of threatening stimuli as a function of
             personality style.},
   Journal = {Brain research bulletin},
   Volume = {74},
   Number = {4},
   Pages = {250-257},
   Year = {2007},
   Month = {September},
   ISSN = {0361-9230},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17720547},
   Abstract = {Cognitive evaluation of emotional stimuli involves a network
             of brain regions including the medial prefrontal cortex
             (mPFC). However, threatening stimuli may be perceived with
             differential salience in different individuals. The goal of
             our study was to evaluate how different personality styles
             are associated with differential modulation of brain
             activity during explicit recognition of fearful and angry
             facial expressions. Twenty-eight healthy subjects underwent
             fMRI. Based on a cognitivist model, subjects were
             categorized according to how they attribute salience to
             emotional stimuli and how they regulate their emotional
             activation. We compared 14 phobic prone (PP) subjects, whose
             identity is more centered on the inner experience ("inward")
             and around control of environmental threat, and 14 eating
             disorders prone (EDP) subjects, whose identity is more
             centered on external referential contexts ("outward") and
             much less around control of threatening stimuli. During fMRI
             subjects either matched the identity of one of two angry and
             fearful faces to that of a simultaneously presented target
             face or identified the expression of a target face by
             choosing one of two simultaneously presented linguistic
             labels. The fMRI results indicated that PP subjects had
             greater mPFC activation when compared with EDP subjects
             during cognitive labeling of threatening stimuli. Activity
             in the mPFC also correlated with personality style scores.
             These results demonstrate that PP subjects recruit greater
             neuronal resources in mPFC whose activity is associated with
             cognitive aspects that are closely intertwined with
             emotional processing. These findings are consistent with the
             contention that cognitive evaluation and salience of
             emotional stimuli are associated with different personality
             styles.},
   Doi = {10.1016/j.brainresbull.2007.06.019},
   Key = {fds251998}
}

@article{Gianaros2007,
   Author = {Gianaros, PJ and Horenstein, JA and Cohen, S and Matthews, KA and Brown,
             SM and Flory, JD and Critchley, HD and Manuck, SB and Hariri,
             AR},
   Title = {Perigenual anterior cingulate morphology covaries with
             perceived social standing.},
   Journal = {Social cognitive and affective neuroscience},
   Volume = {2},
   Number = {3},
   Pages = {161-173},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, PA 15213, USA. gianarospj@upmc.edu},
   Year = {2007},
   Month = {September},
   ISSN = {1749-5016},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/18418472},
   Keywords = {Adult • Cognition/physiology • Female • Gyrus
             Cinguli/*anatomy \& histology/physiology • Humans
             • Male • Middle Aged • *Social Class •
             *Social Perception},
   Abstract = {Low socioeconomic status (SES) increases the risk for
             developing psychiatric and chronic medical disorders. A
             stress-related pathway by which low SES may affect mental
             and physical health is through the perception of holding a
             low social standing, termed low subjective social status.
             This proposal implicates overlapping brain regions mediating
             stress reactivity and socioemotional behaviors as
             neuroanatomical substrates that could plausibly link
             subjective social status to health-related outcomes. In a
             test of this proposal, we used a computational structural
             neuroimaging method (voxel-based morphometry) in a healthy
             community sample to examine the relationships between
             reports of subjective social status and regional gray matter
             volume. Results showed that after accounting for potential
             demographic confounds, subclinical depressive symptoms,
             dispositional forms of negative emotionality and
             conventional indicators of SES, self-reports of low
             subjective social status uniquely covaried with reduced gray
             matter volume in the perigenual area of the anterior
             cingulate cortex (pACC)-a brain region involved in
             experiencing emotions and regulating behavioral and
             physiological reactivity to psychosocial stress. The pACC
             may represent a neuroanatomical substrate by which perceived
             social standing relates to mental and physical
             health.},
   Language = {eng},
   Doi = {10.1093/scan/nsm013},
   Key = {Gianaros2007}
}

@article{Conklin2007,
   Author = {Conklin, SM and Gianaros, PJ and Brown, SM and Yao, JK and Hariri, AR and Manuck, SB and Muldoon, MF},
   Title = {Long-chain omega-3 fatty acid intake is associated
             positively with corticolimbic gray matter volume in healthy
             adults.},
   Journal = {Neuroscience letters},
   Volume = {421},
   Number = {3},
   Pages = {209-212},
   Address = {Department of Psychiatry, University of Pittsburgh, School
             of Medicine, Pittsburgh, PA 15260, United States.
             conklinSM@UPMC.Edu},
   Year = {2007},
   Month = {June},
   ISSN = {0304-3940},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17574755},
   Keywords = {Adult • Brain Mapping • Cerebral Cortex/anatomy \&
             histology/*drug effects • Fatty Acids,
             Omega-3/*administration \& dosage • Female •
             Humans • Limbic System/*anatomy \& histology/*drug
             effects • Magnetic Resonance Imaging/methods •
             Male • Middle Aged},
   Abstract = {<h4>Background</h4>In animals, dendritic arborization and
             levels of brain derived neurotrophic factor are positively
             associated with intake of the omega-3 fatty acids. Here, we
             test whether omega-3 fatty acid intake in humans varies with
             individual differences in gray matter volume, an in vivo,
             systems-level index of neuronal integrity.<h4>Methods</h4>Fifty-five
             healthy adults completed two 24h dietary recall interviews.
             Intake of long-chain omega-3 fatty acids was categorized by
             tertiles. Regional gray matter volumes in a putative
             emotional brain circuitry comprised of the anterior
             cingulate cortex (ACC), amygdala and hippocampus were
             calculated using optimized voxel-based morphometry on
             high-resolution structural magnetic resonance
             images.<h4>Results</h4>Region of interest analyses revealed
             positive associations between reported dietary omega-3
             intake and gray matter volume in the subgenual ACC, the
             right hippocampus and the right amygdala, adjusted for total
             gray matter volume of brain. Unconstrained whole-brain
             analyses confirmed that higher intake of omega-3 fatty acids
             was selectively associated with increased greater gray
             matter volume in these and not other regions.<h4>Conclusions</h4>Higher
             reported consumption of the long-chain omega-3 fatty acids
             is associated with greater gray matter volume in nodes of a
             corticolimbic circuitry supporting emotional arousal and
             regulation. Such associations may mediate previously
             observed effects of omega-3 fatty acids on memory, mood and
             affect regulation.},
   Language = {eng},
   Doi = {10.1016/j.neulet.2007.04.086},
   Key = {Conklin2007}
}

@article{Heinz2007,
   Author = {Heinz, A and Smolka, MN and Braus, DF and Wrase, J and Beck, A and Flor, H and Mann, K and Schumann, G and Büchel, C and Hariri, AR and Weinberger,
             DR},
   Title = {Serotonin transporter genotype (5-HTTLPR): effects of
             neutral and undefined conditions on amygdala
             activation.},
   Journal = {Biological psychiatry},
   Volume = {61},
   Number = {8},
   Pages = {1011-1014},
   Address = {Department of Psychiatry, Charite University Medicine
             Berlin, Campus Charite Mitte, Germany.},
   Year = {2007},
   Month = {April},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17157270},
   Keywords = {Adult • Amygdala/blood supply/*physiology •
             Emotions/*physiology • Functional Laterality •
             Gene Frequency • Genotype • Humans • Image
             Processing, Computer-Assisted/methods • Magnetic
             Resonance Imaging/methods • Male • Middle Aged
             • Oxygen/blood • Photic Stimulation •
             Polymorphism, Genetic • Serotonin Plasma Membrane
             Transport Proteins/*genetics},
   Abstract = {<h4>Background</h4>A polymorphism of the human serotonin
             transporter gene (SCL6A4) has been associated with serotonin
             transporter expression and with processing of aversive
             stimuli in the amygdala. Functional imaging studies show
             that during the presentation of aversive versus neutral
             cues, healthy carriers of the short (s) allele showed
             stronger amygdala activation than long (l) carriers.
             However, a recent report suggested that this interaction is
             driven by amygdala deactivation during presentation of
             neutral stimuli in s carriers.<h4>Methods</h4>Functional MRI
             was used to assess amygdala activation during the
             presentation of a fixation cross or affectively aversive or
             neutral visual stimuli in 29 healthy men.<h4>Results</h4>Amygdala
             activation was increased in s carriers during undefined
             states such as the presentation of a fixation cross compared
             with emotionally neutral conditions.<h4>Conclusions</h4>This
             finding suggests that s carriers show stronger amygdala
             reactivity to stimuli and contexts that are relatively
             uncertain, which we propose are stressful.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2006.08.019},
   Key = {Heinz2007}
}

@article{fds251997,
   Author = {Hariri, AR and Fisher, PM},
   Title = {Regulation of corticolimbic reactivity via the
             5-HT1A autoreceptor in the pathophysiology and
             treatment of depression},
   Journal = {Future Neurology},
   Volume = {2},
   Number = {2},
   Pages = {121-124},
   Publisher = {Future Medicine Ltd},
   Year = {2007},
   Month = {March},
   ISSN = {1479-6708},
   Doi = {10.2217/14796708.2.2.121},
   Key = {fds251997}
}

@article{fds251996,
   Author = {Fisher, PM and Meltzer, CC and Ziolko, SK and Price, JC and Moses-Kolko,
             EL and Berga, SL and Hariri, AR},
   Title = {Erratum: Capacity for 5-HT1A-mediated autoregulation
             predicts amygdala reactivity (Nature Neuroscience (2006) 9
             (1362-1363))},
   Journal = {Nature Neuroscience},
   Volume = {10},
   Number = {2},
   Pages = {263},
   Publisher = {Springer Nature},
   Year = {2007},
   Month = {February},
   ISSN = {1097-6256},
   Doi = {10.1038/nn0207-263a},
   Key = {fds251996}
}

@article{Lerner2007,
   Author = {Lerner, JS and Dahl, RE and Hariri, AR and Taylor,
             SE},
   Title = {Facial expressions of emotion reveal neuroendocrine and
             cardiovascular stress responses.},
   Journal = {Biological psychiatry},
   Volume = {61},
   Number = {2},
   Pages = {253-260},
   Address = {Department of Social \& Decision Sciences, Carnegie Mellon
             University, Pittsburgh, PA 15213, USA. jlerner@cmu.edu},
   Year = {2007},
   Month = {January},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17150197},
   Keywords = {Adolescent • Adult • Anger/physiology •
             Arousal/*physiology • Blood Pressure/*physiology •
             Emotions/*physiology • *Facial Expression • Facial
             Muscles/physiology • Fear/physiology • Female
             • Heart Rate/*physiology • Humans •
             Hydrocortisone/*blood • Hypothalamo-Hypophyseal
             System/*physiology • Male • Pituitary-Adrenal
             System/*physiology • Principal Component Analysis
             • Problem Solving/physiology • Stress,
             Psychological/complications},
   Abstract = {<h4>Background</h4>The classic conception of stress involves
             undifferentiated negative affect and corresponding
             biological reactivity. The present study hypothesized a new
             conception, disaggregating stress into emotion-specific,
             contrasting patterns of biological response. Specifically,
             it hypothesized contrasting patterns for indignation
             (comprised of anger and disgust) versus fear. Moreover, it
             hypothesized that facial expressions of these emotions would
             signal corresponding biological stress responses.<h4>Methods</h4>Ninety-two
             adults engaged in annoyingly difficult stress-challenge
             tasks, during which cardiovascular responses,
             hypothalamic-pituitary-adrenocortical (HPA) axis responses
             (i.e., cortisol), emotional expressions (i.e., facial muscle
             movements), and subjective emotional experience were
             assessed.<h4>Results</h4>Pronounced individual differences
             emerged in specific emotional responses to the stressors.
             Analyses of facial expressions revealed that the more fear
             individuals displayed in response to the stressors, the
             higher their cardiovascular and cortisol responses to
             stress. By contrast, the more indignation individuals
             displayed in response to the same stressors the lower their
             cortisol levels and cardiovascular responses.<h4>Conclusions</h4>Facial
             expressions of emotion signal biological responses to
             stress. Fear expressions signal elevated cortisol and
             cardiovascular reactivity; indignation signals attenuated
             cortisol and cardiovascular reactivity, patterns that
             implicate individual differences in stress appraisals.
             Rather than conceptualizing stress as generalized negative
             affect, studies can be informed by this emotion-specific
             approach to stress responses.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2006.08.016},
   Key = {Lerner2007}
}

@article{fds251995,
   Author = {Bertolino, A and Rubino, V and Sambataro, F and Blasi, G and Latorre, V and Fazio, L and Caforio, G and Petruzzella, V and Kolachana, B and Hariri,
             A and Meyer-Lindenberg, A and Nardini, M and Weinberger, DR and Scarabino, T},
   Title = {Prefrontal-hippocampal coupling during memory processing is
             modulated by COMT val158met genotype.},
   Journal = {Biological psychiatry},
   Volume = {60},
   Number = {11},
   Pages = {1250-1258},
   Year = {2006},
   Month = {December},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16950222},
   Abstract = {<h4>Background</h4>Studies in humans and in animals have
             demonstrated that a network of brain regions is involved in
             performance of declarative and recognition memory tasks.
             This network includes the hippocampal formation (HF) as well
             as the ventrolateral prefrontal cortex (VLPFC). Studies in
             animals have suggested that the relationship between these
             brain regions is strongly modulated by dopamine.<h4>Methods</h4>Using
             fMRI in healthy humans matched for a series of demographic
             and genetic variables, we studied the effect of the COMT
             val158met polymorphism on function of HF and VLPFC as well
             as on their functional coupling during recognition
             memory.<h4>Results</h4>The COMT Val allele was associated
             with: relatively poorer performance at retrieval; reduced
             recruitment of neuronal resources in HF and increased
             recruitment in VLPFC during both encoding and retrieval; and
             unfavorable functional coupling between these two regions at
             retrieval. Moreover, functional coupling during retrieval
             was predictive of behavioral accuracy.<h4>Conclusions</h4>These
             results shed new light on individual differences in
             responsivity and connectivity between HF and VLPFC related
             to genetic modulation of dopamine, a mechanism accounting at
             least in part for individual differences in recognition
             memory performance.},
   Doi = {10.1016/j.biopsych.2006.03.078},
   Key = {fds251995}
}

@article{fds252080,
   Author = {Drabant, EM and Hariri, AR and Meyer-Lindenberg, A and Munoz, KE and Mattay, VS and Kolachana, BS and Egan, MF and Weinberger,
             DR},
   Title = {Catechol O-methyltransferase val158met genotype and neural
             mechanisms related to affective arousal and
             regulation.},
   Journal = {Archives of general psychiatry},
   Volume = {63},
   Number = {12},
   Pages = {1396-1406},
   Year = {2006},
   Month = {December},
   ISSN = {0003-990X},
   Abstract = {<h4>Context</h4>Catechol O-methyltransferase (COMT), the
             major enzyme determining cortical dopamine flux, has a
             common functional polymorphism (val(158)met) that affects
             prefrontal function and working memory capacity and has also
             been associated with anxiety and emotional
             dysregulation.<h4>Objectives</h4>To examine COMT val(158)met
             effects on corticolimbic circuitry reactivity and functional
             connectivity during processing of biologically salient
             stimuli, as well as the relationship to the temperamental
             trait of novelty seeking.<h4>Design</h4>Within-subject
             functional magnetic resonance imaging study.<h4>Setting</h4>National
             Institute of Mental Health, Genes, Cognition, and Psychosis
             Program, Bethesda, Md. Patients One hundred one healthy
             subjects of both sexes.<h4>Results</h4>We found that the met
             allele was associated with a dose-dependent increase in
             hippocampal formation and ventrolateral prefrontal cortex
             activation during viewing of faces displaying negative
             emotion. In met/met homozygotes, limbic and prefrontal
             regions showed increased functional coupling. Moreover, in
             these same subjects, the magnitude of amygdala-orbitofrontal
             coupling was inversely correlated with novelty seeking, an
             index of temperamental inflexibility.<h4>Conclusions</h4>Our
             results indicate that heritable variation in dopamine
             neurotransmission associated with the met allele of the COMT
             polymorphism results in heightened reactivity and
             connectivity in corticolimbic circuits. This may reflect a
             genetic predisposition for inflexible processing of
             affective stimuli, a mechanism possibly accounting for
             aspects of arousal and behavioral control that contribute to
             emotional dysregulation previously reported in met/met
             individuals.},
   Doi = {10.1001/archpsyc.63.12.1396},
   Key = {fds252080}
}

@article{Hariri2006d,
   Author = {Hariri, AR and Brown, SM and Williamson, DE and Flory, JD and de Wit, H and Manuck, SB},
   Title = {Preference for immediate over delayed rewards is associated
             with magnitude of ventral striatal activity.},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {26},
   Number = {51},
   Pages = {13213-13217},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, Pennsylvania 15213, USA. haririar@upmc.edu},
   Year = {2006},
   Month = {December},
   ISSN = {0270-6474},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17182771},
   Keywords = {Adult • Basal Ganglia/*physiology • Behavior,
             Addictive/physiopathology/psychology • Choice
             Behavior/*physiology • Feedback, Psychological/physiology
             • Female • Humans • Impulsive
             Behavior/physiopathology/psychology • Male •
             Middle Aged • Nerve Net/physiology • *Reward
             • Risk Factors • Time Factors},
   Abstract = {Discounting future outcomes as a function of their deferred
             availability underlies much of human decision making.
             Discounting, or preference for immediate over delayed
             rewards of larger value, is often associated with
             impulsivity and is a risk factor for addictive disorders
             such as pathological gambling, cigarette smoking, and drug
             and alcohol abuse. The ventral striatum (VS) is involved in
             mediating behavioral responses and physiological states
             associated with reward, and dysregulation of the VS
             contributes to addiction, perhaps by affecting impulsive
             decision-making. Behavioral tests of delay discounting (DD),
             which index preference for smaller immediate over larger
             delayed rewards, covary with impulsive tendencies in humans.
             In the current study, we examined the relationship between
             individual differences in DD, measured in a behavioral
             assessment, and VS activity measured with blood oxygenation
             level-dependent functional magnetic resonance imaging, in 45
             adult volunteers. VS activity was determined using a task
             involving positive and negative feedback with monetary
             reward. Analyses revealed that individual differences in DD
             correlate positively with magnitude of VS activation in
             response to both positive and negative feedback, compared
             with a no-feedback control condition. Variability in DD was
             also associated with differential VS activation in response
             to positive, compared with negative, feedback. Collectively,
             our results suggest that increased preference for smaller
             immediate over larger delayed rewards reflects both a
             relatively indiscriminate and hyper-reactive VS circuitry.
             They also highlight a specific neurocognitive mechanism that
             may contribute to increased risk for addiction.},
   Language = {eng},
   Doi = {10.1523/jneurosci.3446-06.2006},
   Key = {Hariri2006d}
}

@article{Neumann2006,
   Author = {Neumann, SA and Brown, SM and Ferrell, RE and Flory, JD and Manuck, SB and Hariri, AR},
   Title = {Human choline transporter gene variation is associated with
             corticolimbic reactivity and autonomic-cholinergic
             function.},
   Journal = {Biological psychiatry},
   Volume = {60},
   Number = {10},
   Pages = {1155-1162},
   Address = {Department of Psychiatry and Behavioral Sciences, Eastern
             Virginia Medical School, 825 Fairfax Avenue, Norfolk, VA
             23501. Neumans@evms.edu},
   Year = {2006},
   Month = {November},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16876130},
   Keywords = {Adult • Autonomic Nervous System/*physiology •
             Brain Mapping • Cerebral Cortex/blood
             supply/*physiology • Choice Behavior/physiology •
             Electrocardiography • European Continental Ancestry
             Group • Female • *Genetic Variation • Heart
             Rate/physiology • Humans • Image Processing,
             Computer-Assisted/methods • Limbic System/blood
             supply/*physiology • Magnetic Resonance Imaging/methods
             • Male • Membrane Transport Proteins/*genetics
             • Middle Aged • Neural Pathways/blood
             supply/*physiology • Neuropsychological Tests •
             Oxygen/blood • Polymorphism, Single
             Nucleotide},
   Abstract = {<h4>Background</h4>Our previous work has shown genetic
             variation in the human choline transporter gene (CHT1) to be
             associated with depressive symptoms and autonomic cardiac
             (cholinergic) dysregulation. Here, functional magnetic
             resonance imaging (fMRI) was used to examine the relation
             between a single nucleotide polymorphism (SNP) in CHT1 on
             regional brain reactivity relevant to autonomic
             (cholinergic) function.<h4>Methods</h4>Thirty-two
             participants of European ancestry (18 men, 14 women; age:
             33-54 years) completed an fMRI protocol using corticolimbic
             reactivity and prefrontal inhibitory control paradigms.
             Resting cholinergic function, as measured by heart rate
             variability (HRV), was quantified from electrocardiogram.
             Subjects were genotyped for a CHT1 G/T SNP.<h4>Results</h4>GG
             homozygotes had greater right (R) dorsal amygdala (p <
             .008), bilateral anterior cingulate (p < .009), and R
             caudate reactivity (p < .015) than T-allele carriers. Heart
             rate variability was related to R frontal cortex (Brodmann
             Areas 6, 9, and 46), R hippocampal formation, bilateral
             caudate, and bilateral anterior cingulate reactivity (p's <
             .007).<h4>Conclusions</h4>CHT1 variation is related to
             differences in a distributed corticolimbic circuitry
             mediating behavioral and physiologic arousal. These
             relations may contribute to a biological mechanism by which
             genetic variation in cholinergic neurotransmission affects
             cognition, mood, and autonomic cardiac function.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2006.03.059},
   Key = {Neumann2006}
}

@article{fds252081,
   Author = {Fisher, PM and Meltzer, CC and Ziolko, SK and Price, JC and Moses-Kolko,
             EL and Berga, SL and Hariri, AR},
   Title = {Capacity for 5-HT1A-mediated autoregulation predicts
             amygdala reactivity.},
   Journal = {Nature neuroscience},
   Volume = {9},
   Number = {11},
   Pages = {1362-1363},
   Year = {2006},
   Month = {November},
   ISSN = {1097-6256},
   Abstract = {We examined the contribution of 5-HT1A autoreceptors (with
             [11C]WAY100635 positron emission tomography) to amygdala
             reactivity (with blood oxygenation level-dependent
             functional magnetic resonance imaging) in 20 healthy adult
             volunteers. We found a significant inverse relationship
             wherein 5-HT1A autoreceptor density predicted a notable
             30-44% of the variability in amygdala reactivity. Our data
             suggest a potential molecular mechanism by which a reduced
             capacity for negative feedback regulation of 5-HT release is
             associated with increased amygdala reactivity.},
   Doi = {10.1038/nn1780},
   Key = {fds252081}
}

@article{Hariri2006a,
   Author = {Hariri, AR and Lewis, DA},
   Title = {Genetics and the future of clinical psychiatry.},
   Journal = {The American journal of psychiatry},
   Volume = {163},
   Number = {10},
   Pages = {1676-1678},
   Year = {2006},
   Month = {October},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17012672},
   Keywords = {Forecasting • Genetics, Medical • Mental
             Disorders/genetics • Psychiatry/*trends},
   Language = {eng},
   Doi = {10.1176/ajp.2006.163.10.1676},
   Key = {Hariri2006a}
}

@article{fds251994,
   Author = {Goldberg, TE and Straub, RE and Callicott, JH and Hariri, A and Mattay,
             VS and Bigelow, L and Coppola, R and Egan, MF and Weinberger,
             DR},
   Title = {The G72/G30 gene complex and cognitive abnormalities in
             schizophrenia.},
   Journal = {Neuropsychopharmacology : official publication of the
             American College of Neuropsychopharmacology},
   Volume = {31},
   Number = {9},
   Pages = {2022-2032},
   Year = {2006},
   Month = {September},
   ISSN = {0893-133X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16554747},
   Abstract = {A recently discovered gene complex, G72/G30 (hereafter G72,
             but now termed DAOA), was found to be associated with
             schizophrenia and with bipolar disorder, possibly because of
             an indirect effect on NMDA neurotransmission. In principle,
             if G72 increases risk for psychosis by this mechanism, it
             might impact with greater penetrance those cortically based
             cognitive and neurophysiological functions associated with
             NMDA signaling. We performed two independent family-based
             association studies (one sample contained more than 200
             families and the other more than 65) of multiple SNPs in the
             G72 region and of multiple SNPs in the gene for D-amino acid
             oxidase (DAAO), which may be modulated by G72. We examined
             the relationship between select cognitive measures in
             attention, working memory, and episodic memory and a
             restricted set of G72 SNPs in over 600 normal controls,
             schizophrenic patients, and their nonpsychotic siblings
             using mixed model ANOVAs. We also determined genotype
             effects on neurophysiology measures in normal controls using
             the fMRI BOLD response obtained during activation procedures
             involving either episodic memory or working memory. There
             were no significant single G72 SNP associations and clinical
             diagnosis in either sample, though one approached
             significance (p=0.06). Diagnosis by genotype interaction
             effects for G72 SNP 10 were significant for cognitive
             variables assessing working memory and attention (p=0.05),
             and at the trend level for episodic memory, such that in the
             schizophrenia group an exaggerated allele load effect in the
             predicted directions was observed. In the fMRI paradigms, a
             strong effect of G72 SNP 10 genotype was observed on BOLD
             activation in the hippocampus during the episodic memory
             paradigm. Tests of association with DAAO were consistently
             nonsignificant. We present evidence that SNP variations in
             the G72 gene region increase risk of cognitive impairment in
             schizophrenia. SNP variations were not strongly associated
             with clinical diagnosis in family-based analyses.},
   Doi = {10.1038/sj.npp.1301049},
   Key = {fds251994}
}

@article{Hariri2006c,
   Author = {Hariri, AR and Drabant, EM and Weinberger, DR},
   Title = {Imaging genetics: perspectives from studies of genetically
             driven variation in serotonin function and corticolimbic
             affective processing.},
   Journal = {Biological psychiatry},
   Volume = {59},
   Number = {10},
   Pages = {888-897},
   Address = {Department of Psychiatry, University of Pittsburgh School of
             Medicine, Pittsburgh, Pennsylvania 15213-2593,
             USA.},
   Year = {2006},
   Month = {May},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16442081},
   Keywords = {Alleles • Amygdala/*physiopathology • *Diagnostic
             Imaging • Emotions/*physiology • Genetic
             Variation/*genetics • Humans • Mood
             Disorders/*genetics/physiopathology • Neural
             Pathways/physiopathology • Polymorphism,
             Genetic/*genetics • Prefrontal Cortex/*physiopathology
             • Serotonin/*genetics/physiology • Serotonin
             Plasma Membrane Transport Proteins/genetics • Synaptic
             Transmission/*genetics},
   Abstract = {Advances in molecular biology and neuroimaging have provided
             a unique opportunity to explore the relationships between
             genes, brain, and behavior. In this review, we will briefly
             outline the rationale for studying genetic effects on brain
             function with neuroimaging. We will then use studies of
             genetically driven variation in serotonin transporter
             function on corticolimbic structure and function to
             highlight the effectiveness of this strategy to delineate
             biological pathways and mechanisms by which individual
             differences in brain function emerge and potentially bias
             behavior and risk for psychiatric illness. In a series of
             studies, a relatively frequent regulatory variant of the
             human serotonin transporter gene (5-HTTLPR) has been
             demonstrated to bias the reactivity of the amygdala to
             salient environmental cues. Moreover, the 5-HTTLPR affects
             the development of a broader corticolimbic circuit and
             alters the functional integration of emotional information
             between the amygdala and medial prefrontal cortex. In turn,
             corticolimbic circuit function predicts individual
             differences in an experimental index of temperamental
             anxiety and, thus, might reflect a predictive biological
             marker of increased risk for mood disorders associated with
             the 5-HTTLPR.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2005.11.005},
   Key = {Hariri2006c}
}

@article{Brown2006a,
   Author = {Brown, SM and Manuck, SB and Flory, JD and Hariri,
             AR},
   Title = {Neural basis of individual differences in impulsivity:
             contributions of corticolimbic circuits for behavioral
             arousal and control.},
   Journal = {Emotion (Washington, D.C.)},
   Volume = {6},
   Number = {2},
   Pages = {239-245},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, PA, USA.},
   Year = {2006},
   Month = {May},
   ISSN = {1528-3542},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16768556},
   Keywords = {Adult • Amygdala/*physiopathology •
             Arousal/*physiology • Brain/*physiopathology •
             Cerebral Cortex/*physiopathology • Female • Humans
             • Impulse Control Disorders/diagnosis/*physiopathology
             • Limbic System/*physiopathology • Magnetic
             Resonance Imaging • Male • Middle Aged •
             Nerve Net/*physiology • Neural Inhibition/*physiology
             • Prefrontal Cortex/physiopathology • Severity of
             Illness Index • *Social Control, Informal},
   Abstract = {The objective of the current study was to analyze the neural
             correlates of behavioral arousal and inhibitory control as
             they relate to individual differences in impulsivity via
             well-established functional MRI amygdala reactivity and
             prefrontal inhibitory control paradigms in healthy adult
             subjects. Impulsivity correlated positively with activity of
             the bilateral ventral amygdala, parahippocampal gyrus,
             dorsal anterior cingulate gyrus (BA 32), and bilateral
             caudate. Conversely, impulsivity correlated negatively with
             activity of the dorsal amygdala and ventral prefrontal
             cortex (BA 47). Together, these findings suggest that
             dispositional impulsivity is influenced by the functional
             interplay of corticolimbic behavioral arousal and control
             circuits.},
   Language = {eng},
   Doi = {10.1037/1528-3542.6.2.239},
   Key = {Brown2006a}
}

@article{fds251993,
   Author = {Meyer-Lindenberg, A and Buckholtz, JW and Kolachana, B and R Hariri,
             A and Pezawas, L and Blasi, G and Wabnitz, A and Honea, R and Verchinski,
             B and Callicott, JH and Egan, M and Mattay, V and Weinberger,
             DR},
   Title = {Neural mechanisms of genetic risk for impulsivity and
             violence in humans.},
   Journal = {Proceedings of the National Academy of Sciences of the
             United States of America},
   Volume = {103},
   Number = {16},
   Pages = {6269-6274},
   Year = {2006},
   Month = {April},
   ISSN = {0027-8424},
   Abstract = {Neurobiological factors contributing to violence in humans
             remain poorly understood. One approach to this question is
             examining allelic variation in the X-linked monoamine
             oxidase A (MAOA) gene, previously associated with impulsive
             aggression in animals and humans. Here, we have studied the
             impact of a common functional polymorphism in MAOA on brain
             structure and function assessed with MRI in a large sample
             of healthy human volunteers. We show that the low expression
             variant, associated with increased risk of violent behavior,
             predicted pronounced limbic volume reductions and
             hyperresponsive amygdala during emotional arousal, with
             diminished reactivity of regulatory prefrontal regions,
             compared with the high expression allele. In men, the low
             expression allele is also associated with changes in
             orbitofrontal volume, amygdala and hippocampus
             hyperreactivity during aversive recall, and impaired
             cingulate activation during cognitive inhibition. Our data
             identify differences in limbic circuitry for emotion
             regulation and cognitive control that may be involved in the
             association of MAOA with impulsive aggression, suggest
             neural systems-level effects of X-inactivation in human
             brain, and point toward potential targets for a biological
             approach toward violence.},
   Doi = {10.1073/pnas.0511311103},
   Key = {fds251993}
}

@article{Hariri2006b,
   Author = {Hariri, AR and Holmes, A},
   Title = {Genetics of emotional regulation: the role of the serotonin
             transporter in neural function.},
   Journal = {Trends in cognitive sciences},
   Volume = {10},
   Number = {4},
   Pages = {182-191},
   Address = {Department of Psychiatry and Center for the Neural Basis of
             Cognition, University of Pittsburgh, Pittsburgh,
             Pennsylvania 15213, USA. haririar@upmc.edu},
   Year = {2006},
   Month = {April},
   ISSN = {1364-6613},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16530463},
   Keywords = {Amygdala/pathology/physiology • Animals •
             Emotions/*physiology • *Genetic Variation • Humans
             • Models, Neurological • Mood Disorders/classification/*genetics
             • Serotonin Plasma Membrane Transport
             Proteins/*genetics/metabolism},
   Abstract = {Identifying biological mechanisms through which genes lead
             to individual differences in emotional behavior is paramount
             to our understanding of how such differences confer risk for
             neuropsychiatric illness. The emergence of techniques such
             as in vivo imaging of brain function in humans and genetic
             engineering in rodents has provided important new insights
             into the impact of serotonin (5-HT), a key modulator of
             emotional behavior, on neural systems subserving anxiety and
             depression. A major finding has been the discovery of
             genetic variation in a crucial regulatory molecule within
             the 5-HT system, the 5HT transporter (5-HTT), and its
             influence on emotional traits. The study of the 5-HTT
             provides a new foundation for understanding the
             neurobiological and genetic basis of emotional regulation
             and affective illness.},
   Language = {eng},
   Doi = {10.1016/j.tics.2006.02.011},
   Key = {Hariri2006b}
}

@article{Brown2006b,
   Author = {Brown, SM and Hariri, AR},
   Title = {Neuroimaging studies of serotonin gene polymorphisms:
             exploring the interplay of genes, brain, and
             behavior.},
   Journal = {Cognitive, affective & behavioral neuroscience},
   Volume = {6},
   Number = {1},
   Pages = {44-52},
   Address = {Department of Psychiatry, University of Pittsburgh School of
             Medicine, Western Psychiatric Institute and Clinic, 3811
             O'Hara Street, Room E-729, Pittsburgh, PA 15213-2593,
             USA.},
   Year = {2006},
   Month = {March},
   ISSN = {1530-7026},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16869228},
   Keywords = {Animals • Behavior/*physiology • Brain/*anatomy \&
             histology/*physiology • Brain Mapping • Diagnostic
             Imaging/methods • Emotions/physiology • Humans
             • Nerve Net • *Polymorphism, Genetic •
             Serotonin/*genetics},
   Abstract = {Because of the unique ability it provides to investigate
             information processing at the level of neural systems,
             functional neuroimaging is a powerful tool to explore the
             relationship between genes, brain, and behavior. Recently,
             functional neuroimaging has provided dramatic illustrations
             of how a promoter polymorphism in the human serotonin
             transporter gene, which has been weakly related to several
             dimensions of emotional behaviors (such as neuroticism and
             anxiety traits), is strongly related to the engagement of
             neural systems--namely, the amygdala and subgenual
             prefrontal cortex, subserving emotional information
             processing. This review will outline the experimental
             strategy by which these genetic effects on brain function
             have been explored and highlight the effectiveness of this
             strategy to delineate biological pathways and mechanisms
             contributing to the emergence of individual differences in
             brain function that potentially bias behavior and risk for
             psychiatric illness.},
   Language = {eng},
   Doi = {10.3758/cabn.6.1.44},
   Key = {Brown2006b}
}

@article{Viding2006,
   Author = {Viding, E and Williamson, DE and Hariri, AR},
   Title = {Developmental imaging genetics: challenges and promises for
             translational research.},
   Journal = {Development and psychopathology},
   Volume = {18},
   Number = {3},
   Pages = {877-892},
   Address = {University College London.},
   Year = {2006},
   Month = {January},
   ISSN = {0954-5794},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17152405},
   Keywords = {Antisocial Personality Disorder/genetics/pathology/physiopathology
             • Brain/*pathology/*physiopathology •
             *Electroencephalography • Humans •
             *Interdisciplinary Communication • *Magnetic Resonance
             Imaging • *Mental Disorders/genetics/pathology/physiopathology
             • Research/*standards • Serotonin/genetics},
   Abstract = {Advances in molecular biology, neuroimaging, genetic
             epidemiology, and developmental psychopathology have
             provided a unique opportunity to explore the interplay of
             genes, brain, and behavior within a translational research
             framework. Herein, we begin by outlining an experimental
             strategy by which genetic effects on brain function can be
             explored using neuroimaging, namely, imaging genetics. We
             next describe some major findings in imaging genetics to
             highlight the effectiveness of this strategy for delineating
             biological pathways and mechanisms by which individual
             differences in brain function emerge and potentially bias
             behavior and risk for psychiatric illness. We then discuss
             the importance of applying imaging genetics to the study of
             psychopathology within a developmental framework. By
             beginning to move toward a systems-level approach to
             understanding pathways to behavioral outcomes as they are
             expressed across development, it is anticipated that we will
             move closer to understanding the complexities of the
             specific mechanisms involved in the etiology of psychiatric
             disease. Despite the numerous challenges that lie ahead, we
             believe that developmental imaging genetics has potential to
             yield highly informative results that will ultimately
             translate into public health benefits. We attempt to set out
             guidelines and provide exemplars that may help in designing
             fruitful translational research applications that
             incorporate a developmental imaging genetics
             strategy.},
   Language = {eng},
   Doi = {10.1017/s0954579406060433},
   Key = {Viding2006}
}

@article{Hariri2006e,
   Author = {Hariri, AR and Brown, SM},
   Title = {Serotonin.},
   Journal = {The American journal of psychiatry},
   Volume = {163},
   Number = {1},
   Pages = {12},
   Year = {2006},
   Month = {January},
   ISSN = {0002-953X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16390882},
   Keywords = {Amygdala/*metabolism/physiopathology • European
             Continental Ancestry Group/genetics • Gene Frequency
             • Genetic Variation • Genotype • Humans
             • Mood Disorders/*genetics/physiopathology •
             Serotonin/genetics/metabolism/*physiology • Serotonin
             Plasma Membrane Transport Proteins/*genetics/physiology},
   Language = {eng},
   Doi = {10.1176/appi.ajp.163.1.12},
   Key = {Hariri2006e}
}

@article{Mattay2006,
   Author = {Mattay, VS and Fera, F and Tessitore, A and Hariri, AR and Berman, KF and Das, S and Meyer-Lindenberg, A and Goldberg, TE and Callicott, JH and Weinberger, DR},
   Title = {Neurophysiological correlates of age-related changes in
             working memory capacity.},
   Journal = {Neuroscience letters},
   Volume = {392},
   Number = {1-2},
   Pages = {32-37},
   Address = {Clinical Brain Disorders Branch, Genes, Cognition and
             Psychosis Program, National Institute of Mental Health,
             National Institutes of Health, 9000 Rockville Pike,
             Bethesda, MD 20892, USA. vsm@mail.nih.gov},
   Year = {2006},
   Month = {January},
   ISSN = {0304-3940},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16213083},
   Keywords = {Adult • Age Factors • Aged •
             Aging/*physiology • Drug Combinations • Female
             • Humans • Magnetic Resonance Imaging/methods
             • Male • Memory, Short-Term/*physiology •
             Middle Aged • Neuropsychological Tests/statistics \&
             numerical data • Peroxides/blood • Prefrontal
             Cortex/blood supply/*physiology • Reaction
             Time/physiology • Urea/analogs \& derivatives/blood},
   Abstract = {Cognitive abilities such as working memory (WM) capacity
             decrease with age. To determine the neurophysiological
             correlates of age-related reduction in working memory
             capacity, we studied 10 young subjects (<35 years of age;
             mean age=29) and twelve older subjects (>55 years of age;
             mean age=59) with whole brain blood oxygenation-level
             dependent (BOLD) fMRI on a 1.5 T GE MR scanner using a
             SPIRAL FLASH pulse sequence (TE=24 ms, TR=56 ms, FA=60
             degrees , voxel dimensions=3.75 mm(3)). Subjects performed a
             modified version of the "n" back working memory task at
             different levels of increasing working memory load (1-Back,
             2-Back and 3-Back). Older subjects performed as well as the
             younger subjects at 1-Back (p=0.4), but performed worse than
             the younger subjects at 2-Back (p<0.01) and 3-Back (p=0.06).
             Older subjects had significantly longer reaction time (RT)
             than younger subjects (p<0.04) at all levels of task
             difficulty. Image analysis using SPM 99 revealed a similar
             distribution of cortical activity between younger and older
             subjects at all task levels. However, an analysis of
             variance revealed a significant group x task interaction in
             the prefrontal cortex bilaterally; within working memory
             capacity, as in 1-Back when the older subjects performed as
             well as the younger subjects, they showed greater prefrontal
             cortical (BA 9) activity bilaterally. At higher working
             memory loads, however, when they performed worse then the
             younger subjects, the older subjects showed relatively
             reduced activity in these prefrontal regions. These data
             suggest that, within capacity, compensatory mechanisms such
             as additional prefrontal cortical activity are called upon
             to maintain proficiency in task performance. As cognitive
             demand increases, however, they are pushed past a threshold
             beyond which physiological compensation cannot be made and,
             a decline in performance occurs.},
   Language = {eng},
   Doi = {10.1016/j.neulet.2005.09.025},
   Key = {Mattay2006}
}

@article{Drabant2006,
   Author = {Drabant, E. M. and Hariri, A. R. and Meyer-Lindenberg, A. and Munoz, K. E. and Mattay, V. S. and Kolachana, B. S. and Egan, M. F. and Weinberger, D. R.},
   Title = {Catechol O-methyltransferase val158met genotype and neural
             mechanisms related to affective arousal and
             regulation},
   Journal = {Archives of general psychiatry},
   Volume = {63},
   Number = {12},
   Pages = {1396--406},
   Address = {Genes, Cognition, and Psychosis Program, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health/DHHS, 10 Center Drive, Bethesda, MD
             20892, USA.},
   Year = {2006},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17146014},
   Keywords = {Adult • Affect/*physiology/*radiation effects •
             Affective Symptoms/diagnosis/*genetics •
             Amygdala/physiology • Arousal/genetics/*physiology
             • Brain Mapping/methods • Catechol
             O-Methyltransferase/*genetics • Exploratory
             Behavior/physiology • Facial Expression • Female
             • Genotype • Humans • Limbic
             System/*physiology • Magnetic Resonance
             Imaging/statistics \& numerical data • Male •
             Methionine/genetics • Neural Pathways/physiology •
             Oxygen/blood • Polymorphism, Genetic/*physiology •
             Prefrontal Cortex/*physiology • Synaptic
             Transmission/physiology • Temperament/physiology •
             Valine/genetics • Visual Perception/physiology},
   Abstract = {CONTEXT: Catechol O-methyltransferase (COMT), the major
             enzyme determining cortical dopamine flux, has a common
             functional polymorphism (val(158)met) that affects
             prefrontal function and working memory capacity and has also
             been associated with anxiety and emotional dysregulation.
             OBJECTIVES: To examine COMT val(158)met effects on
             corticolimbic circuitry reactivity and functional
             connectivity during processing of biologically salient
             stimuli, as well as the relationship to the temperamental
             trait of novelty seeking. DESIGN: Within-subject functional
             magnetic resonance imaging study. SETTING: National
             Institute of Mental Health, Genes, Cognition, and Psychosis
             Program, Bethesda, Md. Patients One hundred one healthy
             subjects of both sexes. RESULTS: We found that the met
             allele was associated with a dose-dependent increase in
             hippocampal formation and ventrolateral prefrontal cortex
             activation during viewing of faces displaying negative
             emotion. In met/met homozygotes, limbic and prefrontal
             regions showed increased functional coupling. Moreover, in
             these same subjects, the magnitude of amygdala-orbitofrontal
             coupling was inversely correlated with novelty seeking, an
             index of temperamental inflexibility. CONCLUSIONS: Our
             results indicate that heritable variation in dopamine
             neurotransmission associated with the met allele of the COMT
             polymorphism results in heightened reactivity and
             connectivity in corticolimbic circuits. This may reflect a
             genetic predisposition for inflexible processing of
             affective stimuli, a mechanism possibly accounting for
             aspects of arousal and behavioral control that contribute to
             emotional dysregulation previously reported in met/met
             individuals.},
   Language = {eng},
   Doi = {10.1001/archpsyc.63.12.1396},
   Key = {Drabant2006}
}

@article{Fisher2006,
   Author = {Fisher, P. M. and Meltzer, C. C. and Ziolko, S. K. and Price, J. C. and Moses-Kolko, E. L. and Berga, S. L. and Hariri, A. R.},
   Title = {Capacity for 5-HT1A-mediated autoregulation predicts
             amygdala reactivity},
   Journal = {Nature neuroscience},
   Volume = {9},
   Number = {11},
   Pages = {1362--3},
   Address = {Department of Psychiatry, University of Pittsburgh School of
             Medicine, Pittsburgh, Pennsylvania 15213,
             USA.},
   Year = {2006},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/17013380},
   Keywords = {Adult • Amygdala/*physiology •
             Depression/metabolism • Feedback/physiology •
             Homeostasis/*physiology • Humans • Magnetic
             Resonance Imaging • Oxygen/blood •
             Piperazines/pharmacology • Positron-Emission Tomography
             • Pyridines/pharmacology • Receptor, Serotonin,
             5-HT1A/drug effects/*physiology • Serotonin
             Antagonists/pharmacology},
   Abstract = {We examined the contribution of 5-HT1A autoreceptors (with
             [11C]WAY100635 positron emission tomography) to amygdala
             reactivity (with blood oxygenation level-dependent
             functional magnetic resonance imaging) in 20 healthy adult
             volunteers. We found a significant inverse relationship
             wherein 5-HT1A autoreceptor density predicted a notable
             30-44\% of the variability in amygdala reactivity. Our data
             suggest a potential molecular mechanism by which a reduced
             capacity for negative feedback regulation of 5-HT release is
             associated with increased amygdala reactivity.},
   Language = {eng},
   Doi = {10.1038/nn1780},
   Key = {Fisher2006}
}

@article{fds251992,
   Author = {Fera, F and Weickert, TW and Goldberg, TE and Tessitore, A and Hariri,
             A and Das, S and Lee, S and Zoltick, B and Meeter, M and Myers, CE and Gluck,
             MA and Weinberger, DR and Mattay, VS},
   Title = {Neural mechanisms underlying probabilistic category learning
             in normal aging.},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {25},
   Number = {49},
   Pages = {11340-11348},
   Year = {2005},
   Month = {December},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16339029},
   Abstract = {Probabilistic category learning engages neural circuitry
             that includes the prefrontal cortex and caudate nucleus, two
             regions that show prominent changes with normal aging.
             However, the specific contributions of these brain regions
             are uncertain, and the effects of normal aging have not been
             examined previously in probabilistic category learning. In
             the present study, using a blood oxygenation level-dependent
             functional magnetic resonance imaging block design, 18
             healthy young adults (mean age, 25.5 +/- 2.6 years) and 15
             older adults (mean age, 67.1 +/- 5.3 years) were assessed on
             the probabilistic category learning "weather prediction"
             test. Whole-brain functional images acquired using a 1.5T
             scanner (General Electric, Milwaukee, WI) with gradient
             echo, echo planar imaging (3/1 mm; repetition time, 3000 ms;
             echo time, 50 ms) were analyzed using second-level
             random-effects procedures [SPM99 (Statistical Parametric
             Mapping)]. Young and older adults displayed equivalent
             probabilistic category learning curves, used similar
             strategies, and activated analogous neural networks,
             including the prefrontal and parietal cortices and the
             caudate nucleus. However, the extent of caudate and
             prefrontal activation was less and parietal activation was
             greater in older participants. The percentage correct and
             reaction time were mainly positively correlated with caudate
             and prefrontal activation in young individuals but
             positively correlated with prefrontal and parietal cortices
             in older individuals. Differential activation within a
             circumscribed neural network in the context of equivalent
             learning suggests that some brain regions, such as the
             parietal cortices, may provide a compensatory mechanism for
             healthy older adults in the context of deficient prefrontal
             cortex and caudate nuclei responses.},
   Doi = {10.1523/jneurosci.2736-05.2005},
   Key = {fds251992}
}

@article{fds252011,
   Author = {Dahl, RE and Hariri, AR},
   Title = {Lessons from G. Stanley Hall: Connecting new research in
             biological sciences to the study of adolescent
             development},
   Journal = {Journal of Research on Adolescence},
   Volume = {15},
   Number = {4},
   Pages = {367-382},
   Publisher = {WILEY},
   Year = {2005},
   Month = {December},
   ISSN = {1050-8392},
   Doi = {10.1111/j.1532-7795.2005.00102.x},
   Key = {fds252011}
}

@article{Lerner2005,
   Author = {Lerner, JS and Gonzalez, RM and Dahl, RE and Hariri, AR and Taylor,
             SE},
   Title = {Facial expressions of emotion reveal neuroendocrine and
             cardiovascular stress responses.},
   Journal = {Biological psychiatry},
   Volume = {58},
   Number = {9},
   Pages = {743-750},
   Address = {Department of Social and Decision Sciences, Carnegie Mellon
             University, Pittsburgh, Pennsylvania 15213, USA.
             jlerner@cmu.edu},
   Year = {2005},
   Month = {November},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16256075},
   Keywords = {Adult • *Cardiovascular Physiological Phenomena •
             Emotions/*physiology • *Facial Expression •
             Fear/physiology/psychology • Female •
             Hemodynamics/physiology • Humans •
             Hydrocortisone/blood • Male • Neurosecretory
             Systems/*physiology • Saliva/metabolism • Stress,
             Psychological/*physiopathology},
   Abstract = {<h4>Background</h4>The classic conception of stress involves
             undifferentiated negative affect and corresponding
             biological reactivity. The present study hypothesized a new
             conception that disaggregates stress into emotion-specific,
             contrasting patterns of biological response.<h4>Methods</h4>Ninety-two
             healthy adults engaged in stress-challenge tasks, during
             which cardiovascular responses, hypothalamic-pituitary-adrenocortical
             (HPA) axis responses (i.e., cortisol), emotional expressions
             (i.e., facial muscle movements), and subjective emotional
             experience (self-reported) were assessed.<h4>Results</h4>Pronounced
             individual differences emerged in specific emotional
             responses to the stressors. Analyses of facial expressions
             revealed that the more fear individuals displayed in
             response to the stressors, the higher their cardiovascular
             and cortisol responses to stress. By contrast, the more
             anger and disgust (indignation) individuals displayed in
             response to the same stressors, the lower their cortisol
             levels and cardiovascular responses. Individual differences
             in optimistic appraisals appeared to mediate these
             correlated patterns.<h4>Conclusions</h4>Facial expressions
             of emotion signal biological responses to stress. Fear
             expressions signal elevated cortisol and cardiovascular
             reactivity; anger and disgust signal attenuated cortisol and
             cardiovascular reactivity, patterns that implicate
             individual differences in stress appraisals. Rather than
             conceptualizing stress as generalized negative affect,
             studies can be informed by this emotion-specific approach to
             stress responses.},
   Language = {eng},
   Doi = {10.1016/j.biopsych.2005.08.011},
   Key = {Lerner2005}
}

@article{Brown2005,
   Author = {Brown, SM and Peet, E and Manuck, SB and Williamson, DE and Dahl, RE and Ferrell, RE and Hariri, AR},
   Title = {A regulatory variant of the human tryptophan hydroxylase-2
             gene biases amygdala reactivity},
   Journal = {Molecular Psychiatry},
   Volume = {10},
   Number = {9},
   Pages = {805},
   Publisher = {Springer Nature},
   Address = {Department of Psychiatry, University of Pittsburgh,
             Pittsburgh, PA 15213, USA.},
   Year = {2005},
   Month = {September},
   ISSN = {1359-4184},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16044172},
   Keywords = {Amygdala/*enzymology/physiology • Brain/anatomy \&
             histology/physiology • Emotions/*physiology • Gene
             Frequency • *Genetic Variation • Genotype •
             Humans • Magnetic Resonance Imaging • Oxygen/blood
             • Sequence Deletion • Tryptophan
             Hydroxylase/*genetics},
   Abstract = {Recent studies have indicated that a newly identified second
             isoform of the tryptophan hydroxylase gene (TPH2) is
             preferentially involved in the rate-limiting synthesis of
             neuronal serotonin. Genetic variation in the human TPH2 gene
             (hTPH2) has been associated with altered in vitro enzyme
             activity as well as increased risk for mood disorders. Here,
             we provide the first in vivo evidence that a relatively
             frequent regulatory variant (G(-844)T) of hTPH2 biases the
             reactivity of the amygdala, a neural structure critical in
             the generation and regulation of emotional
             behaviors.},
   Language = {eng},
   Doi = {10.1038/sj.mp.4001725},
   Key = {Brown2005}
}

@article{Lee2005,
   Author = {Lee, M and Bailer, UF and Frank, GK and Henry, SE and Meltzer, CC and Price, JC and Mathis, CA and Putnam, KT and Ferrell, RE and Hariri, AR and Kaye, WH},
   Title = {Relationship of a 5-HT transporter functional polymorphism
             to 5-HT1A receptor binding in healthy women.},
   Journal = {Molecular psychiatry},
   Volume = {10},
   Number = {8},
   Pages = {715-716},
   Year = {2005},
   Month = {August},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15940302},
   Keywords = {Amygdala/physiology • Female • Genotype •
             Humans • Membrane Glycoproteins/*genetics •
             Membrane Transport Proteins/*genetics • Nerve Tissue
             Proteins/*genetics • Neurons/physiology •
             *Polymorphism, Genetic • Receptor, Serotonin,
             5-HT1A/*genetics • Reference Values • Serotonin
             Plasma Membrane Transport Proteins • Statistics,
             Nonparametric},
   Language = {eng},
   Doi = {10.1038/sj.mp.4001680},
   Key = {Lee2005}
}

@article{Meyer-Lindenberg2005,
   Author = {Meyer-Lindenberg, A and Hariri, AR and Munoz, KE and Mervis, CB and Mattay, VS and Morris, CA and Berman, KF},
   Title = {Neural correlates of genetically abnormal social cognition
             in Williams syndrome.},
   Journal = {Nature neuroscience},
   Volume = {8},
   Number = {8},
   Pages = {991-993},
   Address = {Section on Integrative Neuroimaging, National Institute of
             Mental Health, National Institutes of Health, Department of
             Health and Human Services, Bethesda, Maryland 20892, USA.
             andreasml@nih.gov},
   Year = {2005},
   Month = {August},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16007084},
   Keywords = {Amygdala/physiopathology • Anxiety/physiopathology
             • Brain/*physiopathology • Brain Mapping •
             Case-Control Studies • *Cognition • Frontal
             Lobe/physiopathology • Humans • *Magnetic
             Resonance Imaging • Prefrontal Cortex/physiopathology
             • *Social Behavior • Williams Syndrome/*physiopathology/*psychology},
   Abstract = {Williams-Beuren syndrome (WBS), caused by a microdeletion of
             approximately 21 genes on chromosome 7q11.23, is
             characterized by unique hypersociability combined with
             increased non-social anxiety. Using functional neuroimaging,
             we found reduced amygdala activation in individuals with WBS
             for threatening faces but increased activation for
             threatening scenes, relative to matched normal controls.
             Activation and interactions of prefrontal regions linked to
             amygdala, especially orbitofrontal cortex, were abnormal,
             suggesting a genetically controlled neural circuitry for
             regulating human social behavior.},
   Language = {eng},
   Doi = {10.1038/nn1494},
   Key = {Meyer-Lindenberg2005}
}

@article{fds251989,
   Author = {Lieberman, MD and Hariri, A and Jarcho, JM and Eisenberger, NI and Bookheimer, SY},
   Title = {An fMRI investigation of race-related amygdala activity in
             African-American and Caucasian-American individuals.},
   Journal = {Nature neuroscience},
   Volume = {8},
   Number = {6},
   Pages = {720-722},
   Year = {2005},
   Month = {June},
   ISSN = {1097-6256},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15880106},
   Abstract = {Functional magnetic resonance imaging (fMRI) was used to
             examine the nature of amygdala sensitivity to race. Both
             African-American and Caucasian-American individuals showed
             greater amygdala activity to African-American targets than
             to Caucasian-American targets, suggesting that race-related
             amygdala activity may result from cultural learning rather
             than from the novelty of other races. Additionally, verbal
             encoding of African-American targets produced significantly
             less amygdala activity than perceptual encoding of
             African-American targets.},
   Doi = {10.1038/nn1465},
   Key = {fds251989}
}

@article{fds251990,
   Author = {Bertolino, A and Arciero, G and Rubino, V and Latorre, V and De Candia,
             M and Mazzola, V and Blasi, G and Caforio, G and Hariri, A and Kolachana,
             B and Nardini, M and Weinberger, DR and Scarabino,
             T},
   Title = {Variation of human amygdala response during threatening
             stimuli as a function of 5'HTTLPR genotype and personality
             style.},
   Journal = {Biological psychiatry},
   Volume = {57},
   Number = {12},
   Pages = {1517-1525},
   Year = {2005},
   Month = {June},
   ISSN = {0006-3223},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15953488},
   Abstract = {<h4>Background</h4>In the brain, processing of fearful
             stimuli engages the amygdala, and the variability of its
             activity is associated with genetic factors as well as with
             emotional salience. The objective of this study was to
             explore the relevance of personality style for variability
             of amygdala response.<h4>Methods</h4>We studied two groups
             (n=14 in each group) of healthy subjects categorized by
             contrasting cognitive styles with which they attribute
             salience to fearful stimuli: so-called phobic prone subjects
             who exaggerate potential environmental threat versus
             so-called eating disorders prone subjects who tend to be
             much less centered around fear. The two groups underwent
             functional magnetic resonance imaging (fMRI) at 3T during
             performance of a perceptual task of threatening stimuli and
             they were also matched for the genotype of the 5' variable
             number tandem repeat (VNTR) polymorphism in the serotonin
             transporter.<h4>Results</h4>The fMRI results indicated that
             phobic prone subjects selectively recruit the amygdala to a
             larger extent than eating disorders prone subjects. Activity
             in the amygdala was also independently predicted by
             personality style and genotype of the serotonin transporter.
             Moreover, brain activity during a working memory task did
             not differentiate the two groups.<h4>Conclusions</h4>The
             results of the present study suggest that aspects of
             personality style are rooted in biological responses of the
             fear circuitry associated with processing of environmental
             information.},
   Doi = {10.1016/j.biopsych.2005.02.031},
   Key = {fds251990}
}

@article{Pezawas2005,
   Author = {Pezawas, L and Meyer-Lindenberg, A and Drabant, EM and Verchinski,
             BA and Munoz, KE and Kolachana, BS and Egan, MF and Mattay, VS and Hariri,
             AR and Weinberger, DR},
   Title = {5-HTTLPR polymorphism impacts human cingulate-amygdala
             interactions: a genetic susceptibility mechanism for
             depression.},
   Journal = {Nature neuroscience},
   Volume = {8},
   Number = {6},
   Pages = {828-834},
   Address = {Genes, Cognition and Psychosis Program, National Institute
             of Mental Health, National Institutes of Health, 10 Center
             Drive 4S235, Bethesda, Maryland 20892-1379,
             USA.},
   Year = {2005},
   Month = {June},
   ISSN = {1097-6256},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15880108},
   Keywords = {Amygdala/*metabolism/pathology/physiopathology •
             Anthropometry • Anxiety Disorders/*genetics/metabolism/pathology
             • Atrophy/genetics/metabolism/pathology • Brain
             Chemistry/genetics • Brain Mapping • Depressive
             Disorder/*genetics/metabolism/pathology •
             Fear/physiology/psychology • Genetic Predisposition to
             Disease/*genetics • Gyrus Cinguli/*metabolism/pathology/physiopathology
             • Humans • Magnetic Resonance Imaging •
             Membrane Glycoproteins/*genetics • Membrane Transport
             Proteins/*genetics • Mutation/genetics • Nerve
             Tissue Proteins/*genetics • Neural Pathways/metabolism/pathology/physiopathology
             • Neuropsychological Tests • Polymorphism,
             Genetic/genetics • Questionnaires •
             Serotonin/metabolism • Serotonin Plasma Membrane
             Transport Proteins},
   Abstract = {Carriers of the short allele of a functional 5' promoter
             polymorphism of the serotonin transporter gene have
             increased anxiety-related temperamental traits, increased
             amygdala reactivity and elevated risk of depression. Here,
             we used multimodal neuroimaging in a large sample of healthy
             human subjects to elucidate neural mechanisms underlying
             this complex genetic association. Morphometrical analyses
             showed reduced gray matter volume in short-allele carriers
             in limbic regions critical for processing of negative
             emotion, particularly perigenual cingulate and amygdala.
             Functional analysis of those regions during perceptual
             processing of fearful stimuli demonstrated tight coupling as
             a feedback circuit implicated in the extinction of negative
             affect. Short-allele carriers showed relative uncoupling of
             this circuit. Furthermore, the magnitude of coupling
             inversely predicted almost 30% of variation in temperamental
             anxiety. These genotype-related alterations in anatomy and
             function of an amygdala-cingulate feedback circuit critical
             for emotion regulation implicate a developmental,
             systems-level mechanism underlying normal emotional
             reactivity and genetic susceptibility for
             depression.},
   Language = {eng},
   Doi = {10.1038/nn1463},
   Key = {Pezawas2005}
}

@article{Callicott2005,
   Author = {Callicott, JH and Straub, RE and Pezawas, L and Egan, MF and Mattay, VS and Hariri, AR and Verchinski, BA and Meyer-Lindenberg, A and Balkissoon,
             R and Kolachana, B and Goldberg, TE and Weinberger,
             DR},
   Title = {Variation in DISC1 affects hippocampal structure and
             function and increases risk for schizophrenia.},
   Journal = {Proceedings of the National Academy of Sciences of the
             United States of America},
   Volume = {102},
   Number = {24},
   Pages = {8627-8632},
   Address = {Genes, Cognition, and Psychosis Program, Clinical Brain
             Disorders Branch, Division of Intramural Research, National
             Institute of Mental Health, National Institutes of Health,
             Bethesda, MD 20892, USA. callicottj@mail.nih.gov},
   Year = {2005},
   Month = {June},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15939883},
   Keywords = {Adult • African Americans • *Alleles •
             European Continental Ancestry Group • Female •
             Haplotypes/genetics • Hippocampus/*anatomy \&
             histology/metabolism/*physiology • Humans •
             Magnetic Resonance Imaging • Male • Nerve Tissue
             Proteins/*genetics/metabolism • Polymorphism, Single
             Nucleotide/genetics • Psychomotor Performance/physiology
             • Risk Factors • Schizophrenia/*genetics},
   Abstract = {Disrupted-in-schizophrenia 1 (DISC1) is a promising
             schizophrenia candidate gene expressed predominantly within
             the hippocampus. We typed 12 single-nucleotide polymorphisms
             (SNPs) that covered the DISC1 gene. A three-SNP haplotype
             [hCV219779 (C)-rs821597 (G)-rs821616 (A)] spanning 83 kb of
             the gene was associated with schizophrenia in a family-based
             sample (P = 0.002). A common nonconservative SNP (Ser704Cys)
             (rs821616) within this haplotype was associated with
             schizophrenia (P = 0.004). Based on primary expression of
             DISC1 in hippocampus, we hypothesized that allelic variation
             at Ser704Cys would have a measurable impact on hippocampal
             structure and function as assayed via specific
             hippocampus-related intermediate phenotypes. In addition to
             overtransmission in schizophrenia, the Ser allele was
             associated with altered hippocampal structure and function
             in healthy subjects, including reduced hippocampal gray
             matter volume and altered engagement of the hippocampus
             during several cognitive tasks assayed with functional
             magnetic resonance imaging. These convergent data suggest
             that allelic variation within DISC1, either at Ser704Cys or
             haplotypes monitored by it, increases the risk for
             schizophrenia and that the mechanism of this effect involves
             structural and functional alterations in the hippocampal
             formation.},
   Language = {eng},
   Doi = {10.1073/pnas.0500515102},
   Key = {Callicott2005}
}

@article{Tessitore2005,
   Author = {Tessitore, A and Hariri, AR and Fera, F and Smith, WG and Das, S and Weinberger, DR and Mattay, VS},
   Title = {Functional changes in the activity of brain regions
             underlying emotion processing in the elderly.},
   Journal = {Psychiatry research},
   Volume = {139},
   Number = {1},
   Pages = {9-18},
   Address = {Clinical Brain Disorders Branch, Genes, Cognition and
             Psychosis Program, National Institute of Mental Health,
             National Institutes of Health, 10 Center Drive, Bldg. 10,
             Room 3C108, Bethesda, MD 20892-1384, USA.},
   Year = {2005},
   Month = {May},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15936178},
   Keywords = {Adult • *Affect • Aged • Aged, 80 and over
             • Aging/physiology • Amygdala/anatomy \& histology
             • Brain/*anatomy \& histology/*metabolism • Humans
             • *Magnetic Resonance Imaging • Middle Aged •
             Oxygen/metabolism • Prefrontal Cortex/anatomy \&
             histology/metabolism},
   Abstract = {Aging is associated with a decline in both cognitive and
             motor abilities that reflects deterioration of underlying
             brain circuitry. While age-related alterations have also
             been described in brain regions underlying emotional
             behavior (e.g., the amygdala), the functional consequence of
             such changes is less clear. To this end, we used blood
             oxygenation-level dependent (BOLD) functional magnetic
             resonance imaging (fMRI) to explore age-related changes in
             brain regions underlying emotion processing. Twelve young
             (age <30 years) and 14 elderly subjects (age >60 years) were
             studied with BOLD fMRI during a paradigm that involved
             perceptual processing of fearful and threatening stimuli.
             Consistent with previous reports, direct group comparisons
             revealed relatively increased BOLD fMRI responses in
             prefrontal cortical regions, including Broca's area, and
             relatively decreased responses in the amygdala and posterior
             fusiform gyri in elderly subjects. Importantly, additional
             analyses using an elderly-specific brain template for
             spatial normalization of the elderly BOLD fMRI data
             confirmed these divergent regional response patterns. While
             there was no difference between groups in accuracy on the
             task, elderly subjects were significantly slower (delayed
             reaction times) in performing the task. Our current data
             suggest that elderly subjects engage a more distributed
             neocortical network during the perceptual processing of
             emotional facial expressions. In light of recent converging
             data from two other studies, our observed effects may
             reflect age-related compensatory responses and/or
             alternative strategies in processing emotions, as the
             elderly appear to engage cognitive/linguistic systems in the
             context of reduced sensory and/or limbic
             responses.},
   Language = {eng},
   Doi = {10.1016/j.pscychresns.2005.02.009},
   Key = {Tessitore2005}
}

@article{Hariri2005,
   Author = {Hariri, AR and Drabant, EM and Munoz, KE and Kolachana, BS and Mattay,
             VS and Egan, MF and Weinberger, DR},
   Title = {A susceptibility gene for affective disorders and the
             response of the human amygdala.},
   Journal = {Archives of general psychiatry},
   Volume = {62},
   Number = {2},
   Pages = {146-152},
   Address = {Genes, Cognition and Psychosis Program, National Institute
             of Mental Health Intramural Research Program, National
             Institutes of Health, US Department of Health and Human
             Services, Bethesda, MD 20892, USA.},
   Year = {2005},
   Month = {February},
   ISSN = {0003-990X},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15699291},
   Keywords = {Adult • Amygdala/*physiology • Anger/physiology
             • Emotions/*physiology • Facial Expression •
             Fear/physiology • Female • Genetic Predisposition
             to Disease/genetics • Genetic Variation/physiology
             • Genotype • Humans • Magnetic Resonance
             Imaging • Male • Membrane Glycoproteins/*genetics
             • Membrane Transport Proteins/*genetics • Mood
             Disorders/diagnosis/epidemiology/*genetics • Nerve
             Tissue Proteins/*genetics • Oxygen/blood •
             Serotonin Plasma Membrane Transport Proteins • Sex
             Distribution • Stress, Psychological/diagnosis/genetics
             • Transcription, Genetic/physiology • Visual
             Perception/physiology},
   Abstract = {<h4>Background</h4>A common regulatory variant (5-HTTLPR) in
             the human serotonin transporter gene (SLC6A4), resulting in
             altered transcription and transporter availability, has been
             associated with vulnerability for affective disorders,
             including anxiety and depression. A recent functional
             magnetic resonance imaging study suggested that this
             association may be mediated by 5-HTTLPR effects on the
             response bias of the human amygdala-a brain region critical
             for emotional and social behavior-to environmental
             threat.<h4>Objectives and design</h4>To examine the effects
             of 5-HTTLPR genotype on the reactivity of the human amygdala
             to salient environmental cues with functional magnetic
             resonance imaging in a large (N = 92) cohort of volunteers
             carefully screened for past and present medical or
             psychiatric illness, and to explore the effects of 5-HTTLPR
             genotype as well as amygdala reactivity on harm avoidance, a
             putative personality measure related to trait
             anxiety.<h4>Results</h4>We now confirm the finding of
             5-HTTLPR short allele-driven amygdala hyperreactivity in a
             large independent cohort of healthy subjects with no history
             of psychiatric illness or treatment. Furthermore, we
             demonstrate that these genotype effects on amygdala function
             are consistent with a dominant short allele effect and are
             equally prominent in men and women. However, neither
             5-HTTLPR genotype, amygdala reactivity, nor genotype-driven
             variability in this reactivity was reflected in harm
             avoidance scores.<h4>Conclusions</h4>Our results reveal a
             potent modulatory effect of the 5-HTTLPR on amygdala
             reactivity to environmental threat. Since this genetically
             driven effect exists in healthy subjects, it does not, in
             and of itself, predict dimensions of mood or temperament. As
             such, the 5-HTTLPR may represent a classic susceptibility
             factor for affective disorders by biasing the functional
             reactivity of the human amygdala in the context of stressful
             life experiences and/or deficient cortical regulatory
             input.},
   Language = {eng},
   Doi = {10.1001/archpsyc.62.2.146},
   Key = {Hariri2005}
}

@article{Winterer2005,
   Author = {Winterer, G and Hariri, AR and Goldman, D and Weinberger,
             DR},
   Title = {Neuroimaging and human genetics.},
   Journal = {International review of neurobiology},
   Volume = {67},
   Pages = {325-383},
   Address = {Genes, Cognition and Psychosis Program, National Institute
             of Mental Health National Institutes of Health, Bethesda,
             Maryland 20892, USA.},
   Year = {2005},
   Month = {January},
   ISSN = {0074-7742},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/16291027},
   Keywords = {Brain/*anatomy \& histology/physiology • *Brain Mapping
             • *Diagnostic Imaging • *Genetics • Genetics,
             Medical • Humans},
   Abstract = {The past few years have seen a rapid expansion of the
             application of neuroimaging tools to the investigation of
             the genetics of brain structure and function. In this
             chapter, we will (1) highlight the most important steps
             during the historical development of this research field,
             (2) explain the major purposes of using neuroimaging in
             genetic research, (3) address methodological issues that are
             relevant with regard to the application of neuroimaging in
             genetic research, (4) give an overview of the present
             state-of-research, and (5) provide several examples of how
             neuroimaging was successfully applied. © 2005 Elsevier Inc.
             All rights reserved.},
   Language = {eng},
   Doi = {10.1016/s0074-7742(05)67010-9},
   Key = {Winterer2005}
}

@article{Egan2004,
   Author = {Egan, MF and Straub, RE and Goldberg, TE and Yakub, I and Callicott, JH and Hariri, AR and Mattay, VS and Bertolino, A and Hyde, TM and Shannon-Weickert, C and Akil, M and Crook, J and Vakkalanka, RK and Balkissoon, R and Gibbs, RA and Kleinman, JE and Weinberger,
             DR},
   Title = {Variation in GRM3 affects cognition, prefrontal glutamate,
             and risk for schizophrenia.},
   Journal = {Proceedings of the National Academy of Sciences of the
             United States of America},
   Volume = {101},
   Number = {34},
   Pages = {12604-12609},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health/NIH/DHHS,
             Building 10, Center Drive, Bethesda, MD 20892,
             USA.},
   Year = {2004},
   Month = {August},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15310849},
   Keywords = {Adult • Cognition/*physiology • Genetic
             Predisposition to Disease • Genotype •
             Glutamates/*metabolism • Haplotypes •
             Hippocampus/anatomy \& histology/metabolism • Humans
             • Magnetic Resonance Imaging • Middle Aged •
             Neuropsychological Tests • Phenotype • Prefrontal
             Cortex/*metabolism • Receptors, Metabotropic
             Glutamate/genetics/*metabolism • Schizophrenia/*metabolism
             • Sequence Analysis, DNA},
   Abstract = {GRM3, a metabotropic glutamate receptor-modulating synaptic
             glutamate, is a promising schizophrenia candidate gene. In a
             family-based association study, a common GRM3 haplotype was
             strongly associated with schizophrenia (P = 0.0001). Within
             this haplotype, the A allele of single-nucleotide
             polymorphism (SNP) 4 (hCV11245618) in intron 2 was slightly
             overtransmitted to probands (P = 0.02). We studied the
             effects of this SNP on neurobiological traits related to
             risk for schizophrenia and glutamate neurotransmission. The
             SNP4 A allele was associated with poorer performance on
             several cognitive tests of prefrontal and hippocampal
             function. The physiological basis of this effect was
             assessed with functional MRI, which showed relatively
             deleterious activation patterns in both cortical regions in
             control subjects homozygous for the SNP4 A allele. We next
             looked at SNP4's effects on two indirect measures of
             prefrontal glutamate neurotransmission. Prefrontal
             N-acetylaspartate, an in vivo MRI measure related to
             synaptic activity and closely correlated with tissue
             glutamate, was lower in SNP4 AA homozygotes. In postmortem
             human prefrontal cortex, AA homozygotes had lower mRNA
             levels of the glial glutamate transporter EAAT2, a protein
             regulated by GRM3 that critically modulates synaptic
             glutamate. Effects of SNP4 on prefrontal GRM3 mRNA and
             protein levels were marginal. Resequencing revealed no
             missense or splice-site SNPs, suggesting that the intronic
             SNP4 or related haplotypes may exert subtle regulatory
             effects on GRM3 transcription. These convergent data point
             to a specific molecular pathway by which GRM3 genotype
             alters glutamate neurotransmission, prefrontal and
             hippocampal physiology and cognition, and thereby increased
             risk for schizophrenia.},
   Language = {eng},
   Doi = {10.1073/pnas.0405077101},
   Key = {Egan2004}
}

@article{Wang2004,
   Author = {Wang, AT and Dapretto, M and Hariri, AR and Sigman, M and Bookheimer,
             SY},
   Title = {Neural correlates of facial affect processing in children
             and adolescents with autism spectrum disorder.},
   Journal = {Journal of the American Academy of Child and Adolescent
             Psychiatry},
   Volume = {43},
   Number = {4},
   Pages = {481-490},
   Address = {Department of Psychology, UCLA, Los Angeles, CA,
             USA.},
   Year = {2004},
   Month = {April},
   ISSN = {0890-8567},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/15187809},
   Keywords = {Adolescent • Amygdala/*physiopathology • Analysis
             of Variance • Autistic Disorder/*physiopathology •
             Brain Mapping • Case-Control Studies • Cerebral
             Cortex/*physiopathology • Child • Emotions •
             *Facial Expression • Humans • Magnetic Resonance
             Imaging • Male • Psycholinguistics •
             Regression Analysis • *Social Perception},
   Abstract = {<h4>Objective</h4>To examine the neural basis of impairments
             in interpreting facial emotions in children and adolescents
             with autism spectrum disorders (ASD).<h4>Method</h4>Twelve
             children and adolescents with ASD and 12 typically
             developing (TD) controls matched faces by emotion and
             assigned a label to facial expressions while undergoing
             functional magnetic resonance imaging.<h4>Results</h4>Both
             groups engaged similar neural networks during facial emotion
             processing, including activity in the fusiform gyrus (FG)
             and prefrontal cortex. However, between-group analyses in
             regions of interest revealed that when matching facial
             expressions, the ASD group showed significantly less
             activity than the TD group in the FG, but reliably greater
             activity in the precuneus. During the labeling of facial
             emotions, no between-group differences were observed at the
             behavioral or neural level. Furthermore, activity in the
             amygdala was moderated by task demands in the TD group but
             not in the ASD group.<h4>Conclusions</h4>These findings
             suggest that children and adolescents with ASD in part
             recruit different neural networks and rely on different
             strategies when processing facial emotions. High-functioning
             individuals with ASD may be relatively unimpaired in the
             cognitive assessment of basic emotions, yet still show
             differences in the automatic processing of facial
             expressions.},
   Language = {eng},
   Doi = {10.1097/00004583-200404000-00015},
   Key = {Wang2004}
}

@article{Holmes2003,
   Author = {Holmes, A and Hariri, AR},
   Title = {The serotonin transporter gene-linked polymorphism and
             negative emotionality: placing single gene effects in the
             context of genetic background and environment.},
   Journal = {Genes, brain, and behavior},
   Volume = {2},
   Number = {6},
   Pages = {332-335},
   Year = {2003},
   Month = {December},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/14653304},
   Keywords = {Animals • Brain/physiopathology • Carrier
             Proteins/*genetics • Disease Models, Animal •
             Emotions/*physiology • Environment • Humans •
             Membrane Glycoproteins/*genetics • *Membrane Transport
             Proteins • Mice • Mood Disorders/*genetics •
             *Nerve Tissue Proteins • Polymorphism,
             Genetic/*physiology • Promoter Regions,
             Genetic/*genetics • Serotonin • Serotonin Plasma
             Membrane Transport Proteins},
   Language = {eng},
   Doi = {10.1046/j.1601-1848.2003.00052.x},
   Key = {Holmes2003}
}

@article{Hariri2003b,
   Author = {Hariri, AR and Weinberger, DR},
   Title = {Functional neuroimaging of genetic variation in serotonergic
             neurotransmission.},
   Journal = {Genes, brain, and behavior},
   Volume = {2},
   Number = {6},
   Pages = {341-349},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health, US Department of Health and Human
             Services, Bethesda, Maryland, USA. haririar@upmc.edu},
   Year = {2003},
   Month = {December},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/14653306},
   Keywords = {Amygdala/*physiology • Animals • *Brain Mapping
             • Fear/physiology • Genetic Variation •
             Humans • *Magnetic Resonance Imaging • Nerve
             Net/physiology • Neural Pathways/physiology •
             Polymorphism, Genetic/*genetics • Serotonin/*genetics
             • Synaptic Transmission/*genetics},
   Abstract = {Serotonin (5-hydroxytryptamine; 5-HT) is a potent modulator
             of the physiology and behavior involved in generating
             appropriate responses to environmental cues such as danger
             or threat. Furthermore, genetic variation in 5-HT subsystem
             genes can impact upon several dimensions of emotional
             behavior including neuroticism and psychopathology, but
             especially anxiety traits. Recently, functional neuroimaging
             has provided a dramatic illustration of how a promoter
             polymorphism in the human 5-HT transporter (5-HTT) gene,
             which has been weakly related to these behaviors, is
             strongly related to the engagement of neural systems, namely
             the amygdala, subserving emotional processes. In this
             commentary, we discuss how functional neuroimaging can be
             used to characterize the effects of polymorphisms in 5-HT
             subsystem genes on the response of neural circuits
             underlying the generation and regulation of mood and
             temperament as well as susceptibility to affective illness.
             We argue that in time, such knowledge will allow us to not
             only transcend phenomenological diagnosis and represent
             mechanisms of disease, but also identify at-risk individuals
             and biological pathways for the development of new
             treatments.},
   Language = {eng},
   Doi = {10.1046/j.1601-1848.2003.00048.x},
   Key = {Hariri2003b}
}

@article{fds252120,
   Author = {Hariri, AR and Goldberg, TE and Mattay, VS and Kolachana, BS and Callicott, JH and Egan, MF and Weinberger, DR},
   Title = {Brain-derived neurotrophic factor val66met polymorphism
             affects human memory-related hippocampal activity and
             predicts memory performance.},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {23},
   Number = {17},
   Pages = {6690-6694},
   Year = {2003},
   Month = {July},
   Abstract = {BDNF plays a critical role in activity-dependent
             neuroplasticity underlying learning and memory in the
             hippocampus. A frequent single nucleotide polymorphism in
             the targeting region of the human BDNF gene (val66met) has
             been associated with abnormal intracellular trafficking and
             regulated secretion of BDNF in cultured hippocampal neurons
             transfected with the met allele. In addition, the met allele
             has been associated with abnormal hippocampal neuronal
             function as well as impaired episodic memory in human
             subjects, but a direct effect of BDNF alleles on hippocampal
             processing of memory has not been demonstrated. We studied
             the relationship of the BDNF val66met genotype and
             hippocampal activity during episodic memory processing using
             blood oxygenation level-dependent functional magnetic
             resonance imaging and a declarative memory task in healthy
             individuals. Met carriers exhibited relatively diminished
             hippocampal engagement in comparison with val homozygotes
             during both encoding and retrieval processes. Remarkably,
             the interaction between the BDNF val66met genotype and the
             hippocampal response during encoding accounted for 25% of
             the total variation in recognition memory performance. These
             data implicate a specific genetic mechanism for substantial
             normal variation in human declarative memory and suggest
             that the basic effects of BDNF signaling on hippocampal
             function in experimental animals are important in
             humans.},
   Doi = {10.1523/jneurosci.23-17-06690.2003},
   Key = {fds252120}
}

@article{fds252077,
   Author = {Mattay, VS and Goldberg, TE and Fera, F and Hariri, AR and Tessitore, A and Egan, MF and Kolachana, B and Callicott, JH and Weinberger,
             DR},
   Title = {Catechol O-methyltransferase val158-met genotype and
             individual variation in the brain response to
             amphetamine.},
   Journal = {Proceedings of the National Academy of Sciences of the
             United States of America},
   Volume = {100},
   Number = {10},
   Pages = {6186-6191},
   Year = {2003},
   Month = {May},
   ISSN = {0027-8424},
   Abstract = {Monamines subserve many critical roles in the brain, and
             monoaminergic drugs such as amphetamine have a long history
             in the treatment of neuropsychiatric disorders and also as a
             substance of abuse. The clinical effects of amphetamine are
             quite variable, from positive effects on mood and cognition
             in some individuals, to negative responses in others,
             perhaps related to individual variations in monaminergic
             function and monoamine system genes. We explored the effect
             of a functional polymorphism (val(158)-met) in the catechol
             O-methyltransferase gene, which has been shown to modulate
             prefrontal dopamine in animals and prefrontal cortical
             function in humans, on the modulatory actions of amphetamine
             on the prefrontal cortex. Amphetamine enhanced the
             efficiency of prefrontal cortex function assayed with
             functional MRI during a working memory task in subjects with
             the high enzyme activity val/val genotype, who presumably
             have relatively less prefrontal synaptic dopamine, at all
             levels of task difficulty. In contrast, in subjects with the
             low activity met/met genotype who tend to have superior
             baseline prefrontal function, the drug had no effect on
             cortical efficiency at low-to-moderate working memory load
             and caused deterioration at high working memory load. These
             data illustrate an application of functional neuroimaging in
             pharmacogenomics and extend basic evidence of an
             inverted-"U" functional-response curve to increasing
             dopamine signaling in the prefrontal cortex. Further,
             individuals with the met/met catechol O-methyltransferase
             genotype appear to be at increased risk for an adverse
             response to amphetamine.},
   Doi = {10.1073/pnas.0931309100},
   Key = {fds252077}
}

@article{Hariri2003c,
   Author = {Hariri, AR and Mattay, VS and Tessitore, A and Fera, F and Weinberger,
             DR},
   Title = {Neocortical modulation of the amygdala response to fearful
             stimuli.},
   Journal = {Biological psychiatry},
   Volume = {53},
   Number = {6},
   Pages = {494-501},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health, Department of Health and Human
             Services, Bethesda, Maryland 20892, USA.},
   Year = {2003},
   Month = {March},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12644354},
   Keywords = {Adult • Affect • Amygdala/*anatomy \&
             histology/*metabolism • Cognition/physiology •
             Equipment Design • *Fear • Female • Galvanic
             Skin Response/physiology • Humans • Magnetic
             Resonance Imaging/instrumentation • Male •
             Oxygen/blood • Photic Stimulation • Prefrontal
             Cortex/*anatomy \& histology/*metabolism},
   Abstract = {<h4>Background</h4>The cortical circuitry involved in
             conscious cognitive processes and the subcortical circuitry
             involved in fear responses have been extensively studied
             with neuroimaging, but their interactions remain largely
             unexplored. A recent functional magnetic resonance imaging
             (fMRI) study demonstrated that the engagement of the right
             prefrontal cortex during the cognitive evaluation of angry
             and fearful facial expressions is associated with an
             attenuation of the response of the amygdala to these same
             stimuli, providing evidence for a functional neural network
             for emotional regulation.<h4>Methods</h4>In the current
             study, we have explored the generalizability of this
             functional network by using threatening and fearful non-face
             stimuli derived from the International Affective Picture
             System (IAPS), as well as the influence of this network on
             peripheral autonomic responses.<h4>Results</h4>Similar to
             the earlier findings with facial expressions, blood oxygen
             level dependent fMRI revealed that whereas perceptual
             processing of IAPS stimuli was associated with a bilateral
             amygdala response, cognitive evaluation of these same
             stimuli was associated with attenuation of this amygdala
             response and a correlated increase in response of the right
             prefrontal cortex and the anterior cingulate cortex.
             Moreover, this pattern was reflected in changes in skin
             conductance.<h4>Conclusions</h4>The current results further
             implicate the importance of neocortical regions, including
             the prefrontal and anterior cingulate cortices, in
             regulating emotional responses mediated by the amygdala
             through conscious evaluation and appraisal.},
   Language = {eng},
   Doi = {10.1016/s0006-3223(02)01786-9},
   Key = {Hariri2003c}
}

@article{Hariri2003a,
   Author = {Hariri, AR and Weinberger, DR},
   Title = {Imaging genomics.},
   Journal = {British medical bulletin},
   Volume = {65},
   Pages = {259-270},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health, Bethesda, Maryland 20892,
             USA.},
   Year = {2003},
   Month = {January},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12697630},
   Keywords = {Apolipoproteins E/genetics • Brain/*pathology •
             Brain Diseases/*genetics/pathology • Carrier
             Proteins/genetics • Catechol O-Methyltransferase/genetics
             • *Genomics • Humans • *Magnetic Resonance
             Imaging • Membrane Glycoproteins/genetics •
             *Membrane Transport Proteins • *Nerve Tissue Proteins
             • Polymorphism, Genetic • Serotonin Plasma
             Membrane Transport Proteins},
   Abstract = {The recent completion of a working draft of the human genome
             sequence promises to provide unprecedented opportunities to
             explore the genetic basis of individual differences in
             complex behaviours and vulnerability to neuropsychiatric
             illness. Functional neuroimaging, because of its unique
             ability to assay information processing at the level of
             brain within individuals, provides a powerful approach to
             such functional genomics. Recent fMRI studies have
             established important physiological links between functional
             genetic polymorphisms and robust differences in information
             processing within distinct brain regions and circuits that
             have been linked to the manifestation of various disease
             states such as Alzheimer's disease, schizophrenia and
             anxiety disorders. Importantly, all of these biological
             relationships have been revealed in relatively small samples
             of healthy volunteers and in the absence of observable
             differences at the level of behaviour, underscoring the
             power of a direct assay of brain physiology like fMRI in
             exploring the functional impact of genetic
             variation.},
   Language = {eng},
   Doi = {10.1093/bmb/65.1.259},
   Key = {Hariri2003a}
}

@article{Hariri2003d,
   Author = {Hariri, A. R. and Goldberg, T. E. and Mattay, V. S. and Kolachana, B. S. and Callicott, J. H. and Egan, M. F. and Weinberger, D. R.},
   Title = {Brain-derived neurotrophic factor val66met polymorphism
             affects human memory-related hippocampal activity and
             predicts memory performance},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {23},
   Number = {17},
   Pages = {6690--4},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health, United States Department of Health and
             Human Services, Bethesda, Maryland 20892-1384,
             USA.},
   Year = {2003},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12890761},
   Keywords = {Adult • Amino Acid Substitution/*genetics •
             Brain/anatomy \& histology/physiology • Brain Mapping
             • Brain-Derived Neurotrophic Factor/*genetics •
             Cohort Studies • DNA Mutational Analysis • Female
             • Genotype • Heterozygote •
             Hippocampus/anatomy \& histology/*physiology •
             Homozygote • Humans • Magnetic Resonance Imaging
             • Male • Memory/*physiology • Photic
             Stimulation • Polymorphism, Genetic/*genetics •
             Reference Values • Regression Analysis},
   Abstract = {BDNF plays a critical role in activity-dependent
             neuroplasticity underlying learning and memory in the
             hippocampus. A frequent single nucleotide polymorphism in
             the targeting region of the human BDNF gene (val66met) has
             been associated with abnormal intracellular trafficking and
             regulated secretion of BDNF in cultured hippocampal neurons
             transfected with the met allele. In addition, the met allele
             has been associated with abnormal hippocampal neuronal
             function as well as impaired episodic memory in human
             subjects, but a direct effect of BDNF alleles on hippocampal
             processing of memory has not been demonstrated. We studied
             the relationship of the BDNF val66met genotype and
             hippocampal activity during episodic memory processing using
             blood oxygenation level-dependent functional magnetic
             resonance imaging and a declarative memory task in healthy
             individuals. Met carriers exhibited relatively diminished
             hippocampal engagement in comparison with val homozygotes
             during both encoding and retrieval processes. Remarkably,
             the interaction between the BDNF val66met genotype and the
             hippocampal response during encoding accounted for 25\% of
             the total variation in recognition memory performance. These
             data implicate a specific genetic mechanism for substantial
             normal variation in human declarative memory and suggest
             that the basic effects of BDNF signaling on hippocampal
             function in experimental animals are important in
             humans.},
   Language = {eng},
   Key = {Hariri2003d}
}

@article{Mattay2003,
   Author = {Mattay, V. S. and Goldberg, T. E. and Fera, F. and Hariri,
             A. R. and Tessitore, A. and Egan, M. F. and Kolachana, B. and Callicott, J. H. and Weinberger, D. R.},
   Title = {Catechol O-methyltransferase val158-met genotype and
             individual variation in the brain response to
             amphetamine},
   Journal = {Proceedings of the National Academy of Sciences of the
             United States of America},
   Volume = {100},
   Number = {10},
   Pages = {6186--91},
   Address = {Clinical Brain Disorders Branch, National Institute of
             Mental Health, National Institutes of Health, Department of
             Health and Human Services, Building 10, Center Drive, Room
             4S-235, Bethesda, MD 20982-1379, USA.},
   Year = {2003},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12716966},
   Keywords = {Amino Acid Substitution • Brain/drug
             effects/enzymology/*physiology • Catechol
             O-Methyltransferase/*genetics • DNA/genetics/isolation
             \& purification • Dextroamphetamine/*pharmacology
             • Genotype • Humans • Magnetic Resonance
             Imaging • Methionine • Organ Specificity •
             Valine},
   Abstract = {Monamines subserve many critical roles in the brain, and
             monoaminergic drugs such as amphetamine have a long history
             in the treatment of neuropsychiatric disorders and also as a
             substance of abuse. The clinical effects of amphetamine are
             quite variable, from positive effects on mood and cognition
             in some individuals, to negative responses in others,
             perhaps related to individual variations in monaminergic
             function and monoamine system genes. We explored the effect
             of a functional polymorphism (val(158)-met) in the catechol
             O-methyltransferase gene, which has been shown to modulate
             prefrontal dopamine in animals and prefrontal cortical
             function in humans, on the modulatory actions of amphetamine
             on the prefrontal cortex. Amphetamine enhanced the
             efficiency of prefrontal cortex function assayed with
             functional MRI during a working memory task in subjects with
             the high enzyme activity val/val genotype, who presumably
             have relatively less prefrontal synaptic dopamine, at all
             levels of task difficulty. In contrast, in subjects with the
             low activity met/met genotype who tend to have superior
             baseline prefrontal function, the drug had no effect on
             cortical efficiency at low-to-moderate working memory load
             and caused deterioration at high working memory load. These
             data illustrate an application of functional neuroimaging in
             pharmacogenomics and extend basic evidence of an
             inverted-``U'' functional-response curve to increasing
             dopamine signaling in the prefrontal cortex. Further,
             individuals with the met/met catechol O-methyltransferase
             genotype appear to be at increased risk for an adverse
             response to amphetamine.},
   Language = {eng},
   Doi = {10.1073/pnas.0931309100},
   Key = {Mattay2003}
}

@article{fds251988,
   Author = {Tessitore, A and Hariri, AR and Fera, F and Smith, WG and Chase, TN and Hyde, TM and Weinberger, DR and Mattay, VS},
   Title = {Erratum: Dopamine modulates the response of the human
             amygdala: A study in Parkinson's disease (Journal of
             Neuroscience (October 15, 2002) (9099-9103))},
   Journal = {Journal of Neuroscience},
   Volume = {22},
   Number = {24},
   Pages = {i},
   Year = {2002},
   Month = {December},
   Key = {fds251988}
}

@article{Hariri2002c,
   Author = {Hariri, AR and Mattay, VS and Tessitore, A and Fera, F and Smith, WG and Weinberger, DR},
   Title = {Dextroamphetamine modulates the response of the human
             amygdala.},
   Journal = {Neuropsychopharmacology : official publication of the
             American College of Neuropsychopharmacology},
   Volume = {27},
   Number = {6},
   Pages = {1036-1040},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health, Bethesda, MD, USA.
             hariria@intra.nimh.nih.gov},
   Year = {2002},
   Month = {December},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12464460},
   Keywords = {Adult • Amygdala/*drug effects/physiology •
             Dextroamphetamine/*pharmacology • Double-Blind Method
             • Emotions/*drug effects/physiology • Facial
             Expression • Female • Humans • Magnetic
             Resonance Imaging/methods/statistics \& numerical data
             • Male},
   Abstract = {Amphetamine, a potent monoaminergic agonist, has pronounced
             effects on emotional behavior in humans, including the
             generation of fear and anxiety. Recent animal studies have
             demonstrated the importance of monoamines, especially
             dopamine, in modulating the response of the amygdala, a key
             brain region involved in the perception of fearful and
             threatening stimuli, and the generation of appropriate
             physiological and behavioral responses. We have explored the
             possibility that the anxiogenic effect of amphetamine in
             humans reflects the drug's influence on the activity of the
             amygdala. In a double-blind placebo controlled study, fMRI
             revealed that dextroamphetamine potentiated the response of
             the amygdala during the perceptual processing of angry and
             fearful facial expressions. Our results provide the first
             evidence of a specific neural substrate for the anxiogenic
             effects of amphetamine and are consistent with animal models
             of dopaminergic activation of the amygdala.},
   Language = {eng},
   Doi = {10.1016/s0893-133x(02)00373-1},
   Key = {Hariri2002c}
}

@article{Tessitore2002,
   Author = {Tessitore, A and Hariri, AR and Fera, F and Smith, WG and Chase, TN and Hyde, TM and Weinberger, DR and Mattay, VS},
   Title = {Dopamine modulates the response of the human amygdala: a
             study in Parkinson's disease.},
   Journal = {The Journal of neuroscience : the official journal of the
             Society for Neuroscience},
   Volume = {22},
   Number = {20},
   Pages = {9099-9103},
   Address = {Clinical Brain Disorders Branch, National Institute of
             Mental Health, National Institutes of Health, Bethesda,
             Maryland 20892-1384, USA.},
   Year = {2002},
   Month = {October},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12388617},
   Keywords = {Amygdala/drug effects/*physiopathology • Antiparkinson
             Agents/therapeutic use • Behavior • Brain Mapping
             • Dopamine/*metabolism/pharmacology • Dopamine
             Agonists/therapeutic use • Drug Therapy, Combination
             • Emotions/physiology • Facial Expression •
             Female • Humans • Magnetic Resonance Imaging
             • Male • Middle Aged • Parkinson
             Disease/*physiopathology • Pattern Recognition, Visual
             • Photic Stimulation • Reaction Time •
             Reference Values},
   Abstract = {In addition to classic motor signs and symptoms, Parkinson's
             disease (PD) is characterized by neuropsychological and
             emotional deficits, including a blunted emotional response.
             In the present study, we explored both the neural basis of
             abnormal emotional behavior in PD and the physiological
             effects of dopaminergic therapy on the response of the
             amygdala, a central structure in emotion processing. PD
             patients and matched normal controls (NCs) were studied with
             blood oxygenation level-dependent functional magnetic
             resonance imaging during a paradigm that involved perceptual
             processing of fearful stimuli. PD patients were studied
             twice, once during a relatively hypodopaminergic state
             (i.e., > or =12 hr after their last dose of dopamimetic
             treatment) and again during a dopamine-replete state. The
             imaging data revealed a robust bilateral amygdala response
             in NCs that was absent in PD patients during the
             hypodopaminergic state. Dopamine repletion partially
             restored this response in PD patients. Our results
             demonstrate an abnormal amygdala response in PD that may
             underlie the emotional deficits accompanying the disease.
             Furthermore, consistent with findings in experimental animal
             paradigms, our results provide in vivo evidence of the role
             of dopamine in modulating the response of the amygdala to
             sensory information in human subjects.},
   Language = {eng},
   Doi = {10.1523/jneurosci.22-20-09099.2002},
   Key = {Tessitore2002}
}

@article{Hariri2002a,
   Author = {Hariri, AR and Tessitore, A and Mattay, VS and Fera, F and Weinberger,
             DR},
   Title = {The amygdala response to emotional stimuli: a comparison of
             faces and scenes.},
   Journal = {NeuroImage},
   Volume = {17},
   Number = {1},
   Pages = {317-323},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health, Bethesda, Maryland 20892, USA.
             hariria@intra.nimh.nih.gov},
   Year = {2002},
   Month = {September},
   ISSN = {1053-8119},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12482086},
   Keywords = {Adult • Amygdala/anatomy \& histology/*physiology
             • Anger/physiology • Emotions/*physiology •
             *Facial Expression • Fear/physiology • Female
             • Galvanic Skin Response/physiology • Humans
             • Image Interpretation, Computer-Assisted •
             Individuality • Magnetic Resonance Imaging • Male
             • Reaction Time/physiology • Social
             Perception},
   Abstract = {As a central fear processor of the brain, the amygdala
             initiates a cascade of critical physiological and behavioral
             responses. Neuroimaging studies have shown that the human
             amygdala responds not only to fearful and angry facial
             expressions but also to fearful and threatening scenes such
             as attacks, explosions, and mutilations. Given the relative
             importance of facial expressions in adaptive social
             behavior, we hypothesized that the human amygdala would
             exhibit a stronger response to angry and fearful facial
             expressions in comparison to other fearful and threatening
             stimuli. Twelve subjects completed two tasks while
             undergoing fMRI: matching angry or fearful facial
             expressions, and matching scenes depicting fearful or
             threatening situations derived from the International
             Affective Picture System (IAPS). While there was an amygdala
             response to both facial expressions and IAPS stimuli, direct
             comparison revealed that the amygdala response to facial
             expressions was significantly greater than that to IAPS
             stimuli. Autonomic reactivity, measured by skin conductance
             responses, was also greater to facial expressions. These
             results suggest that the human amygdala shows a stronger
             response to affective facial expressions than to scenes, a
             bias that should be considered in the design of experimental
             paradigms interested in probing amygdala
             function.},
   Language = {eng},
   Doi = {10.1006/nimg.2002.1179},
   Key = {Hariri2002a}
}

@article{Hariri2002b,
   Author = {Hariri, AR and Mattay, VS and Tessitore, A and Kolachana, B and Fera, F and Goldman, D and Egan, MF and Weinberger, DR},
   Title = {Serotonin transporter genetic variation and the response of
             the human amygdala.},
   Journal = {Science (New York, N.Y.)},
   Volume = {297},
   Number = {5580},
   Pages = {400-403},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, National
             Institutes of Health, Bethesda, MD 20892,
             USA.},
   Year = {2002},
   Month = {July},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/12130784},
   Keywords = {Adult • Alleles • Amygdala/*physiology •
             Carrier Proteins/*genetics/physiology • Cohort Studies
             • Facial Expression • *Fear • Female •
             Functional Laterality • *Genetic Variation •
             Genotype • Humans • Magnetic Resonance Imaging
             • Male • Membrane Glycoproteins/*genetics/physiology
             • *Membrane Transport Proteins • *Nerve Tissue
             Proteins • Personality • Polymorphism, Genetic
             • *Promoter Regions, Genetic •
             Serotonin/*metabolism • Serotonin Plasma Membrane
             Transport Proteins • Sex Characteristics},
   Abstract = {A functional polymorphism in the promoter region of the
             human serotonin transporter gene (SLC6A4) has been
             associated with several dimensions of neuroticism and
             psychopathology, especially anxiety traits, but the
             predictive value of this genotype against these complex
             behaviors has been inconsistent. Serotonin [5-
             hydroxytryptamine, (5-HT)] function influences normal fear
             as well as pathological anxiety, behaviors critically
             dependent on the amygdala in animal models and in clinical
             studies. We now report that individuals with one or two
             copies of the short allele of the serotonin transporter
             (5-HTT) promoter polymorphism, which has been associated
             with reduced 5-HTT expression and function and increased
             fear and anxiety-related behaviors, exhibit greater amygdala
             neuronal activity, as assessed by BOLD functional magnetic
             resonance imaging, in response to fearful stimuli compared
             with individuals homozygous for the long allele. These
             results demonstrate genetically driven variation in the
             response of brain regions underlying human emotional
             behavior and suggest that differential excitability of the
             amygdala to emotional stimuli may contribute to the
             increased fear and anxiety typically associated with the
             short SLC6A4 allele.},
   Language = {eng},
   Doi = {10.1126/science.1071829},
   Key = {Hariri2002b}
}

@article{Mattay2002,
   Author = {Mattay, VS and Fera, F and Tessitore, A and Hariri, AR and Das, S and Callicott, JH and Weinberger, DR},
   Title = {Neurophysiological correlates of age-related changes in
             human motor function.},
   Journal = {Neurology},
   Volume = {58},
   Number = {4},
   Pages = {630-635},
   Address = {Clinical Brain Disorders Branch, Intramural Research
             Program, National Institute of Mental Health, NIH, Bethesda,
             MD 20892-1379, USA. vsm@helix.nih.gov},
   Year = {2002},
   Month = {February},
   ISSN = {0028-3878},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/11865144},
   Keywords = {Adult • Aging/*physiology • Analysis of Variance
             • Brain/*physiology • Female • Humans •
             Magnetic Resonance Imaging/statistics \& numerical data
             • Male • Middle Aged • Psychomotor
             Performance/*physiology},
   Abstract = {<h4>Background</h4>There are well-defined and characteristic
             age-related deficits in motor abilities that may reflect
             structural and chemical changes in the aging
             brain.<h4>Objective</h4>To delineate age-related changes in
             the physiology of brain systems subserving simple motor
             behavior.<h4>Methods</h4>Ten strongly right-handed young
             (<35 years of age) and 12 strongly right-handed elderly (>50
             years of age) subjects with no evidence of cognitive or
             motor deficits participated in the study. Whole-brain
             functional imaging was performed on a 1.5-T MRI scanner
             using a spiral pulse sequence while the subjects performed a
             visually paced "button-press" motor task with their dominant
             right hand alternating with a rest state.<h4>Results</h4>Although
             the groups did not differ in accuracy, there was an increase
             in reaction time in the elderly subjects (mean score plus
             minus SD, young subjects = 547 +/- 97 ms, elderly subjects =
             794 +/- 280 ms, p < 0.03). There was a greater extent of
             activation in the contralateral sensorimotor cortex, lateral
             premotor area, supplementary motor area, and ipsilateral
             cerebellum in the elderly subjects relative to the young
             subjects (p < 0.001). Additional areas of activation, absent
             in the young subjects, were seen in the ipsilateral
             sensorimotor cortex, putamen (left > right), and
             contralateral cerebellum of the elderly subjects.<h4>Conclusions</h4>The
             results of this study show that elderly subjects recruit
             additional cortical and subcortical areas even for the
             performance of a simple motor task. These changes may
             represent compensatory mechanisms invoked by the aging
             brain, such as reorganization and redistribution of
             functional networks to compensate for age-related structural
             and neurochemical changes.},
   Language = {eng},
   Doi = {10.1212/wnl.58.4.630},
   Key = {Mattay2002}
}

@article{fds252009,
   Author = {Hariri, A and Sigman, M and Bookheimer, S},
   Title = {Abnormal regional brain activity associated with processing
             emotional stimuli in autistic subjects},
   Journal = {NeuroImage},
   Volume = {11},
   Number = {5 PART II},
   Pages = {S242},
   Year = {2000},
   Month = {January},
   Doi = {10.1016/s1053-8119(00)91174-1},
   Key = {fds252009}
}

@article{Hariri2000,
   Author = {Hariri, AR and Bookheimer, SY and Mazziotta, JC},
   Title = {Modulating emotional responses: effects of a neocortical
             network on the limbic system.},
   Journal = {Neuroreport},
   Volume = {11},
   Number = {1},
   Pages = {43-48},
   Address = {Brain Mapping Division, UCLA School of Medicine,
             Ahmanson-Lovelace Brain Mapping Center 90095,
             USA.},
   Year = {2000},
   Month = {January},
   ISSN = {0959-4965},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/10683827},
   Keywords = {Adult • Amygdala/blood supply/physiology •
             Anger/physiology • Cerebrovascular Circulation/physiology
             • Emotions/*physiology • Facial Expression •
             Fear/physiology • Female • Humans • Image
             Interpretation, Computer-Assisted • Limbic System/blood
             supply/*physiology • Magnetic Resonance Imaging •
             Male • Neocortex/blood supply/*physiology • Nerve
             Net/*physiology • Social Perception},
   Abstract = {Humans share with animals a primitive neural system for
             processing emotions such as fear and anger. Unlike other
             animals, humans have the unique ability to control and
             modulate instinctive emotional reactions through
             intellectual processes such as reasoning, rationalizing, and
             labeling our experiences. This study used functional MRI to
             identify the neural networks underlying this ability.
             Subjects either matched the affect of one of two faces to
             that of a simultaneously presented target face (a perceptual
             task) or identified the affect of a target face by choosing
             one of two simultaneously presented linguistic labels (an
             intellectual task). Matching angry or frightened expressions
             was associated with increased regional cerebral blood flow
             (rCBF) in the left and right amygdala, the brain's primary
             fear centers. Labeling these same expressions was associated
             with a diminished rCBF response in the amygdalae. This
             decrease correlated with a simultaneous increase in rCBF in
             the right prefrontal cortex, a neocortical region implicated
             in regulating emotional responses. These results provide
             evidence for a network in which higher regions attenuate
             emotional responses at the most fundamental levels in the
             brain and suggest a neural basis for modulating emotional
             experience through interpretation and labeling.},
   Language = {eng},
   Doi = {10.1097/00001756-200001170-00009},
   Key = {Hariri2000}
}

@article{Hale2000,
   Author = {Hale, TS and Hariri, AR and McCracken, JT},
   Title = {Attention-deficit/hyperactivity disorder: perspectives from
             neuroimaging},
   Journal = {Mental retardation and developmental disabilities research
             reviews},
   Volume = {6},
   Number = {3},
   Pages = {214-219},
   Address = {UCLA School of Medicine, Los Angeles, California 90024,
             USA.},
   Year = {2000},
   ISSN = {1080-4013},
   url = {http://www.ncbi.nlm.nih.gov/pubmed/10982499},
   Keywords = {Attention Deficit Disorder with Hyperactivity/*pathology/physiopathology
             • Brain/*pathology/radiography/radionuclide imaging
             • Electroencephalography • Humans • Magnetic
             Resonance Imaging • Tomography, Emission-Computed,
             Single-Photon • Tomography, X-Ray Computed},
   Abstract = {Attention-deficit/hyperactivity disorder (ADHD) is a common
             childhood behavioral disorder most often characterized by
             inattentiveness, impulsivity, and hyperactivity. Current
             etiologic theories suggest that ADHD stems from
             abnormalities in dopaminergic and possibly noradrenergic
             brainstem nuclei that act to regulate a cortico-striato-thalamo-cortical
             network, believed to be critical for executive functions and
             the regulation of behavioral responses such as arousal,
             attention, and inhibition. Noninvasive structural and
             functional neuroimaging techniques provide a new avenue for
             exploring the validity of these theories. Despite their
             limitations, neuroimaging studies have provided a unique and
             otherwise inaccessible, biological perspective of ADHD.
             Collectively, the studies reviewed here reveal that
             dysfunction in arousal, behavioral inhibition, and attention
             associated with ADHD may result from structural
             abnormalities in frontostriatal regions that in turn result
             in diminished activity essential for normal
             function.},
   Language = {eng},
   Doi = {10.1002/1098-2779(2000)6:33.0.CO;2-M},
   Key = {Hale2000}
}

@article{fds252007,
   Author = {Dapretto, M and Hariri, A and Bialik, MH and Bookheimer,
             SY},
   Title = {Cortical correlates of affective vs. linguistic prosody: An
             fMRI study},
   Journal = {NeuroImage},
   Volume = {9},
   Number = {6 PART II},
   Pages = {S1054},
   Year = {1999},
   Month = {December},
   ISSN = {1053-8119},
   Key = {fds252007}
}

@article{fds252008,
   Author = {Hariri, A and Quintana, J and Kovalik, E and Bookheimer,
             S},
   Title = {Processing of facial affect in schizophrenics: An fMRI
             study},
   Journal = {NeuroImage},
   Volume = {9},
   Number = {6 PART II},
   Pages = {S647},
   Year = {1999},
   Month = {December},
   ISSN = {1053-8119},
   Key = {fds252008}
}

@article{fds252006,
   Author = {Hariri, AR and Werren, JH and Wilkinson, GS},
   Title = {Distribution and reproductive effects of Wolbachia in
             stalk-eyed flies (Diptera: Diopsidae)},
   Journal = {Heredity},
   Volume = {81},
   Number = {3},
   Pages = {254-260},
   Publisher = {Springer Nature America, Inc},
   Year = {1998},
   Month = {September},
   Abstract = {Wolbachia are cytoplasmically inherited bacteria capable of
             altering the reproductive biology of their hosts in a manner
             which increases their spread within a population. These
             microbes can cause cytoplasmic incompatibility,
             parthenogenesis and feminization of genetic males. Because
             Wolbachia have been associated with female-biased sex ratio
             distortion, we used a PCR assay to examine 17 species of
             stalk-eyed flies (Diptera: Diopsidae), two of which exhibit
             female-biased sex ratios, for the presence of these
             microbes. Type A Wolbachia was detected in four diopsid
             species, three from the genus Sphyracephala, none of which
             exhibit biased progeny sex ratios. The reproductive effects
             of the microbe were examined in one of those species, S.
             beccarii, by conducting reciprocal crosses between infected
             and uninfected strains. In this species, Wolbachia do not
             cause detectable cytoplasmic incompatibility or reduce host
             fecundity. In contrast, our results are consistent with an
             association between the microbes and enhanced male
             fertility. Possible explanations for the pattern of
             distribution and effects on male fertility include a
             predisposition for acquiring Type A Wolbachia by these flies
             and accommodation by the host genome to bacterial
             presence.},
   Doi = {10.1038/sj.hdy.6883640},
   Key = {fds252006}
}

@article{fds252005,
   Author = {Bookheimer, SY and Zeineh, MM and Strojwas, MH and Hariri,
             AR},
   Title = {Localization of memory-related activation on a picture
             priming task using fMRI},
   Journal = {NeuroImage},
   Volume = {7},
   Number = {4 PART II},
   Pages = {S824},
   Year = {1998},
   Month = {January},
   Doi = {10.1016/s1053-8119(18)31657-4},
   Key = {fds252005}
}