ArticlePDF Available

Reproducible Genetic Risk Loci for Anxiety: Results From ∼200,000 Participants in the Million Veteran Program

Authors:

Abstract and Figures

Objective: Anxiety disorders are common and often disabling. The goal of this study was to examine the genetic architecture of anxiety disorders and anxiety symptoms, which are also frequently comorbid with other mental disorders, such as major depressive disorder. Methods: Using one of the world's largest biobanks including genetic, environmental, and medical information, the Million Veteran Program, the authors performed a genome-wide association study (GWAS) of a continuous trait for anxiety (based on score on the Generalized Anxiety Disorder 2-item scale [GAD-2], N=199,611) as the primary analysis and self-report of physician diagnosis of anxiety disorder (N=224,330) as a secondary analysis. Results: The authors identified five genome-wide significant signals for European Americans and one for African Americans on GAD-2 score. The strongest were on chromosome 3 (rs4603973) near SATB1, a global regulator of gene expression, and on chromosome 6 (rs6557168) near ESR1, which encodes an estrogen receptor. The locus identified on chromosome 7 (rs56226325, MAF=0.17) near MAD1L1 was previously identified in GWASs of bipolar disorder and schizophrenia. The authors replicated these findings in the summary statistics of two major published GWASs for anxiety, and also found evidence of significant genetic correlation between the GAD-2 score results and previous GWASs for anxiety (rg=0.75), depression (rg=0.81), and neuroticism (rg=0.75). Conclusions: This is the largest GWAS of anxiety traits to date. The authors identified novel genome-wide significant associations near genes involved with global regulation of gene expression (SATB1) and the estrogen receptor alpha (ESR1). Additionally, the authors identified a locus (MAD1L1) that may have implications for genetic vulnerability across several psychiatric disorders. This work provides new insights into genetic risk mechanisms underpinning anxiety and related psychiatric disorders.
Content may be subject to copyright.
Reproducible Genetic Risk Loci for Anxiety: Results From
200,000 Participants in the Million Veteran Program
Daniel F. Levey, Ph.D., Joel Gelernter, M.D., Renato Polimanti, Ph.D., Hang Zhou, Ph.D., Zhongshan Cheng, Ph.D.,
Mihaela Aslan, Ph.D., Rachel Quaden, M.A., John Concato, M.D., M.P.H., Krishnan Radhakrishnan, M.D., Ph.D.,
Julien Bryois, Ph.D., Patrick F. Sullivan, M.D., the Million Veteran Program, Murray B. Stein, M.D., M.P.H.
Objective: Anxiety disorders are common and often dis-
abling. The goal of this study was to examine the genetic
architecture of anxiety disorders and anxiety symptoms,
which are also frequently comorbid with other mental dis-
orders, such as major depressive disorder.
Methods: Using one of the worlds largest biobanks including
genetic, environmental, and medical information, the Million
Veteran Program, the authors performed a genome-wide
association study (GWAS) of a continuous trait for anxiety
(based on score on the Generalized Anxiety Disorder 2-item
scale [GAD-2], N=199,611) as the primary analysis and self-
report of physician diagnosis of anxiety disorder (N=224,330)
as a secondary analysis.
Results: The authors identied ve genome-wide signicant
signals for European Americans and one for African Ameri-
cans on GAD-2 score. The strongest were on chromosome
3 (rs4603973) near SATB1, a global regulator of gene ex-
pression, and on chromosome 6 (rs6557168) near ESR1,
which encodes an estrogen receptor. The locus identied
on chromosome 7 (rs56226325, MAF=0.17) near MAD1L1
was previously identied in GWASs of bipolar disorder and
schizophrenia. The authors replicated these ndings in the
summary statistics of two major published GWASs for anxiety,
and also found evidence of signicant genetic correlation
between the GAD-2 score results and previous GWASs for
anxiety (r
g
=0.75), depression (r
g
=0.81), and neuroticism
(r
g
=0.75).
Conclusions: This is the largest GWAS of anxiety traits to date.
The authors identied novel genome-wide signicant as-
sociations near genes involved with global regulation of gene
expression (SATB1) and the estrogen receptor alpha (ESR1).
Additionally, the authors identied a locus (MAD1L1) that may
have implications for genetic vulnerability across several
psychiatric disorders. This work provides new insights into
genetic risk mechanisms underpinning anxiety and related
psychiatric disorders.
AJP in Advance (doi: 10.1176/appi.ajp.2019.19030256)
Anxiety disorders are common, affecting 1 in 10 Americans
each year, and are a leading cause of disability worldwide (1).
An analysis of health expenditures in the United States found
that anxiety and depressive disorders together accounted for
about $90 billion in personal health spending in the United
States in 2013 (2). Given their prevalence, associated im-
pairment, and economic costs, anxiety disorders are a major
public health concern (3).
Anxiety is a future-oriented mood state associated with
preparation for possible, upcoming negative events(4) and is
usually a normal and adaptive behavioral response to ev-
eryday life. In anxiety disorders, anxiety is excessive or out of
proportion to the actual or anticipated event and is accom-
panied by clinically signicant distress or disability (5).
Numerous risk factors for anxiety disorders have been
studied, including experiential and genetic factors (6). For
example, neurotic personality traits are predictive of the
onset of anxiety disorders (7). Twin studies demonstrate
a heritable component to anxiety disorders (6), but there
have been few published genome-wide association studies
(GWASs) to date investigating anxiety or anxiety-related
traits. The Anxiety Neuro Genetics Study (ANGST) (8)
meta-analysis was the rst large GWAS to identify signicant
genetic associations, nding one genome-wide signicant
locus each for a categorical case-control design for any
anxiety disorder diagnosis and a quantitative factor score for
anxiety in a cohort of over 18,000 subjects. Another recent
large GWAS, from the Lundbeck Foundation Initiative for
Integrative Psychiatric Research (iPSYCH), identied a
signicant genetic association with anxiety and stress-related
disorders in a cohort of 31,880 individuals in the national
Danish registers (9). Also of note is a study based on the UK
Biobank cohort, the second largest GWAS of anxiety to date
(10), which examined anxiety using case-control and clinical
ajp in Advance ajp.psychiatryonline.org 1
ARTICLES
cutoffs based on a score $10 on the Generalized Anxiety
Disorder 7-item scale (GAD-7). While progress is being made,
understanding of the genetics of anxiety disorders has lagged
behind other related disorders, such as major depression (11).
Only a third of individuals with anxiety disorders receive
treatment (12). For those who do enter treatment, psycho-
logical approaches such as cognitive-behavioral therapy
(CBT) have been shown to be effective (13), as have certain
pharmacotherapies (14). A recent systematic review of CBT
treatment response rates for anxiety disorders showed av-
erage rates of 49.5% at end of treatment and 53.6% at follow-
up (15). A better understanding of genetic risk factors and
determinants now informs other aspects of medicine, such as
oncology and cardiology, through identication of causal
mutations (16) and variants, and this approach will have
important implications for psychiatry (17). These precision-
medicine approaches are challenging in complex traits such
as anxiety, which are associated with many (perhaps hun-
dreds of thousands of ) variants of individually small effect
(18). The use of polygenic risk scores will require a suitably
large sample size to provide sufcient condence in these
small individual effects that cumulatively account for so
much of the heritability (19). Underlying polygenic risk
factors from sufciently large cohorts may inform an ap-
proach to identifying individuals with a predisposition to
anxiety disorders and to improving outcomes.
The Million Veteran Program (MVP), one of the worlds
largest biobanks including genetic, environmental, and
medical information, is based on data from U.S. military
veterans (2022). Using this large genetic data set and the
Generalized Anxiety Disorder 2-item scale (GAD-2) (23) as
well as self-reported physician diagnosis of an anxiety dis-
order, we discovered novel genome-wide signicant variants
associated with anxiety in European Americans and African
Americans. We examined replication and genetic overlap of
these results with previous studies of anxiety and traits with
which anxiety disorders are commonly comorbidmajor
depression, PTSD, and neuroticism. We also examined ex-
pression quantitative trait loci (eQTLs) to identify possible
gene expression implications of these genetic variants, with
eQTL evidence for altered expression in the basal ganglia and
cerebellum. These ndings, in the largestcohort of individuals
analyzed by GWAS for anxiety and anxiety disorders (199,611
subjects for the quantitative trait, 224,330 for binary self-
report diagnosis) to date, indicate shared genetic risk with
some other mental disorders but also point to loci that may be
especially important for anxiety and anxiety-related traits.
METHODS
Participants
The MVP cohort has been described previously (20). Results
were analyzed in two separate tranches based on when geno-
typing results were available. Ancestry was assigned using
10 principal components and the 1000 Genomes Project phase
3 EUR and AFR data as reference within each MVP tranche.
Genotyping, Imputation, and Quality Control
Genotyping, imputation, and quality control within MVP has
been previously described. Briey, samples were genotyped
using a 723,305-SNP Affymetrix Axiom Biobank array, cus-
tomized for MVP (20). Imputation was performed with
minimac3 using data from the 1000 Genomes Project. For
postimputation quality control, SNPs with an imputation
INFO score ,0.3 or a minor allele frequency (MAF) ,0.001
were removed from analysis. For the rst tranche of data,
22,183 SNPs were selected through linkage disequilibrium
(LD) pruning using PLINK 2.0 (24), and then Eigensoft (25)
was used to conduct principal component analysis on 343,286
MVP samples and 2,504 1000 Genomes samples. The ref-
erence population groups (EUR, EAS, AFR, AMR, or SAS) in
the 1000 Genomes samples were used to dene European
American (N=241,541) and African American (N=61,796)
groups used in this analysis. Similar methods were used in the
second tranche of data, which contained 108,416 new MVP
samples and the same 2,504 1000 Genomes samples. In
tranche 2, 80,694 participants were dened as European
American and 20,584 as African American.
Phenotypic Assessment
We used the GAD-2 (23) for our primary analysis. The GAD-2
consists of two questions (see Table S1 in the online sup-
plement) in a self-report survey, each scored on a scale of 03.
Participants are asked to respond according to their symp-
toms during the past 2 weeks. Values for the two responses
are summed, resulting in a range of scores between 0 and 6,
which we treated as a continuous trait (Table 1). Mean GAD-2
scores in European American men (N=163,470) and women
(N=11,693) were 1.08 (SD=1.64) and 1.64 (SD=1.87), re-
spectively, and mean scores in African American men
(N=21,153) and women (N=3,295) were 1.57 (SD=10.21) and
1.94 (SD=10.64), respectively. The mean ages of the European
American and African American participants were 66.58
years (SD=11.62) and 60.6 years (SD=10.78), respectively.
Another anxiety phenotypeself-reported physician di-
agnosis of anxiety disorderwas analyzed based on data
collected from the MVP baseline survey. Participants were
asked, Please tell us if you have been diagnosed with the
following conditions: anxiety reaction/panic disorder.An-
swers were recorded as yes/no binary responses, and missing
responses were excluded from analysis. A total of 224,330
participants (34,189 case subjects who responded yes, 190,141
control subjects who responded no) had available phenotype
and genotype information and had assignments of either
European ancestry (28,525 cases, 163,731 controls) or African
ancestry (5,664 cases, 26,410 controls) (see Table S2 in the
online supplement).
Statistical Analysis
GWAS analysis was carried out by linear regression for each
ancestry group and tranche using PLINK 2.0 on geno-
type dosage data, covarying for age, sex, and the rst 10 prin-
cipal components against the phenotype of GAD-2 score.
2ajp.psychiatryonline.org ajp in Advance
REPRODUCIBLE GENETIC RISK LOCI FOR ANXIETY
Ancestry-specicandtrans-ancestry meta-analysis were per-
formed using inverse variance weighting in the METAL software
package (European American, N=175,163; African American,
N=24,448; combined trans-ancestry, N=199,611). Logistic re-
gression was used for self-reported physician diagnosis of an
anxiety disorder, and the results obtained were combined using
the same meta-analytic approach. To identify independent
GWAS signals, we clumped results using an r
2
of 0.10 and
window size of 1,000 kb. Post-GWAS analyses were conducted
for what turned out to be the most genetically informative
phenotype based on z-scored heritability: GAD-2 score.
Linkage Disequilibrium Score Regression
and SNP-Based Heritability
We used linkage disequilibrium score regression through
LD Hub (26) to estimate SNP-based heritability and to
assess genetic correlation of GAD-2 anxiety with all traits
available in LD Hub. The traits from the ANGST GWAS of
anxiety case-control and factor scores (8) and iPSYCH
anxiety and stress-related disorders (9)neither of which
were available in LD Hubwere calculated separately
with LD score regression software (LDSC) using sum-
mary statistics downloaded from the Psychiatric Geno-
mics Consortium (PGC) web site (https://www.med.
unc.edu/pgc/results-and-downloads/) or from the authors,
respectively.
Conditional Analysis for Major Depression
Considering the extensive comorbidity between major de-
pression and anxiety disorders (6), we ran a conditional
analysis with the multi-trait-based conditional and joint
analysis method (mtCOJO) (27) using the GCTA software
package. This method uses GWAS summary statistics from
one trait to perform conditional analysis on GWAS summary
statistics from another trait. We conditioned the MVP GAD-2
summary statistics as the primary analysis with the PGC
major depressive disorder (11) summary statistics for de-
pression. We quantied changes in variance explained by
using LD score regression to calculate heritability in the
depression-conditioned GAD-2 analysis and compared with
the original GAD-2 analysis.
Gene-Based Tests
Summary statistics from the
GWAS were loaded into the
FUMA (Functional Map-
ping and Annotation) GWAS
platform to test for gene-level
associations using Multi-
Marker Analysis of Geno-
Mic Annotation (MAGMA)
(28). Input SNPs were
mapped to 18,469 protein
coding genes. The genome-
wide signicance thresh-
oldforthegene-basedtest
was dened in accordance
with Bonferroni multiple testing correction (p=0.05/
18,469=2.71310
26
).
Fine Mapping
Fine mapping was conducted using PAINTOR, version 3 (29).
A brain functional annotation set (30) was used to prioritize
causal SNPs. The z-scored GAD-2 GWAS summary statistics
served as the base analysis data set, with the aforementioned
brain data set serving as the functional annotation. We
enumerated all possible combinations and searched for a
single causal SNP within each locus.
RESULTS
Primary Analysis
GWAS of GAD-2 scores was conducted separately in two
tranches of each ancestry in the MVP sample, dened by the
time when data became available, and meta-analyzed to-
gether within ancestral group. One genomic locus was
genome-wide signicant in the African American meta-
analysis (Figure 1A), and ve loci were genome-wide sig-
nicant in the European American meta-analysis (Figure 1B).
The genome-wide signicant result from the African
American analysis (rs575403075, MAF=0.06, p=2.82310
28
)
was near the TRPV6 (Transient Receptor Potential Cation
Channel Subfamily V Member 6) locus. The top signal in the
European American meta-analysis consisted of 64 genome-
wide signicant SNPs in high LD at the SATB1-AS1 (Special
AT-Rich Sequence Binding 1 Antisense RNA 1) locus on
chromosome 3. The strongest nding (rs4603973, MAF=0.29,
p=6.18310
211
) was intronic at SATB1-AS1.Thesecond
strongest independent signal was on chromosome
6 (rs6557168, MAF=0.37, p=1.33310
29
) intronic at ESR1
(Estrogen Receptor 1) with 10 other genome-wide signicant
SNPs in high LD. A third genome-wide signicant association
for European Americans was found on chromosome
1 (rs12023347, MAF=0.48, p=8.88310
29
) near the long
noncoding RNA LINC01360 and LRRIQ3 (Leucine-Rich
Repeats and IQ motif containing 3). The fourth genome-
wide signicant association found in European Americans
was on chromosome 7 (rs56226325, MAF=0.17, p=2.01310
28
)
TABLE 1. Subjects and phenotype distribution in a study of genetic risk loci for anxiety
a
European Americans African Americans
Age Age
GAD-2 Score N Mean SE Female (%) N Mean SE Female (%)
0 100,141 69.10 0.033 4.79 11,212 62.93 0.1 10.64
1 21,569 65.31 0.082 8.20 2,897 60.33 0.2 15.15
2 25,061 63.95 0.075 8.86 3,947 58.97 0.166 16.06
3 8,450 62.40 0.129 9.44 1,708 58.86 0.243 14.87
4 8,046 61.49 0.134 10.08 1,730 57.83 0.253 16.99
5 4,447 60.61 0.176 9.83 1,011 57.30 0.333 15.23
6 7,449 59.16 0.143 11.63 1,943 56.40 0.235 16.83
Total 175,163 66.58 0.028 6.68 24,448 60.59 0.069 13.48
a
GAD-2=Generalized Anxiety Disorder 2-item scale.
ajp in Advance ajp.psychiatryonline.org 3
LEVEY ET AL.
in an intron of MAD1L1 (Mitotic Arrest Decient 1 Like 1). The
fth association for European Americans was on chromosome
20 in and around the TCEA2 (Transcription Elongation Factor
A2), RGS19 (Regulator of G Protein Signaling 19), and OPRL1
(Opioid Related Nociceptin Receptor 1) genes (rs6090040,
MAF=0.48, p=3.28310
28
).
We conducted additional analyses using case-control
status for self-reported physician diagnosis of an anxiety
disorder. For the European American subjects, there were
two genome-wide signicant signals for the GWAS of self-
reported physician diagnosis of an anxiety disorder, in a gene-
rich region nearest AURKB on chromosome 17 (rs35546597,
MAF=0.42, p=1.88310
28
) and on chromosome 7 in an IQCE
intron (rs10534613, MAF=0.41, p=4.92310
28
) close to the
MAD1L1 locus identied for GAD-2. There were no genome-
wide signicant ndings for this phenotype in African
Americans.
Replication
For replication, wetested our top ve SNPsfrom the analysis of
GAD-2 scores in European Americans in three independent
GWASs with anxiety-related phenotypes. We considered a
replication to be signicant if the p value was ,0.05. We in-
vestigated our lead genome-wide signicant SNPs in GWASs
for the ANGST anxiety case-control (8), iPSYCH anxiety and
stress-related disorders (9), and UK Biobank, 23andMe, and
Genetics of Personality Consortium (GPC) neuroticism (31)
phenotypes (Table 2). The rst two phenotypes are very
similar to our GAD-2 measure; the third is best considered a
related phenotype (r
g
=0.7174, p=1.95310
253
).
In the ANGST anxiety
study (8), which was the
smallest replication cohort,
all ve of our top in-
dependent SNPs had the
same direction of effect, with
two being nominally signi-
cant (p,0.05). In the
iPSYCH study of anxiety and
stress-related disorders (9),
four of ve independent
SNPs had the same direction
of effect, with three being
nominally signicant (p,0.05).
We also replicated the lead
SNP from iPSYCH near
PDE4B in our own study
(iPSYCH lead SNP: rs7528604,
p=5.39310
28
; present study
GAD-2: same SNP, p=0.015).
Only one of our ndings, near
OPRL1, failed to replicate in at
least one independent study.
Our lead SNP on chro-
mosome 3 near the SATB1
locus, rs4603973, was not
available for lookup in the neuroticism GWAS (31), which we
used as a proxy replication of a related trait, so we used the
strongest LD-proxy SNP available (rs4390955 R
2
=0.91,
p=7.78E-11). In this study, four of our ve independent SNPs
we looked up had the same direction of effect, three were
nominally signicant (p,0.05), and one near MAD1L1 was
nearly genome-wide signicant (UK Biobank neuroticism:
rs56226325, p=6.59310
28
; present study GAD-2: same SNP,
p=2.01310
28
).
Lastly, a preprint reported results for anxiety from the
UK Biobank using case-controlandtheGAD-7,scoredasa
dichotomous trait (10). We found signicant replication
fortwooftheirfourndings, with suggestive evidence
for a third (see Table S9 in the online supplement).
Genome-Wide Gene-Based Association Study for GAD-2
In the genome-wide gene-based association study, the top
gene identied was OPRL1 (p=1.15310
29
), which was also
signicant in the SNP-wise analysis, as noted above. Thirty-
one genes were identied as genome-wide signicant fol-
lowing Bonferroni correction for multiple comparisons. A
more permissive Benjamini-Hochberg correction performed
by step-up procedure, with genes ranked by p value and
corrected for 18,469 individual tests with a still relatively
restrictive 0.05 false discovery rate, identied 189 genes (see
Table S3 in the online supplement) in total for investigation
of biological relevance through the Ingenuity pathway en-
richment tool (32), Ingenuity Pathway Analysis (Ingenuity
Systems, Redwood City, Calif.; www.ingenuity.com) (see
Table S4 in the online supplement). Among the top enriched
FIGURE 1. Circle Manhattan plot for anxiety phenotypes in African Americans and European
Americans
a
a
The outer circle displays results of the Generalized Anxiety Disorder 2-item scale genome-wide association
study (GWAS), and the inner circle contains results for the case-control (self-report of physician diagnosis of
anxiety disorder) GWAS. Numbers outside the circle represent chromosomes. Red dots indicate genome-wide
signicant ndings (p,5 x10
28
) and yellow dots indicate suggestive ndings (p,5310
26
). The scale on the
y-axis represents 2log
10
(p value). Vertical dashed gray lines are drawn through genome-wide signicant
ndings to indicate overlap between analyses. The genes nearest to the lead SNP are labeled adjacent to the
result. In most cases a genome-wide signicant (red) locus from one phenotype overlaps with at least a
suggestive (yellow) locus in the other.
4ajp.psychiatryonline.org ajp in Advance
REPRODUCIBLE GENETIC RISK LOCI FOR ANXIETY
diseases or functional annotations were carcinoma (p=1.76310
27
)
and fear conditioning (p=3.62310
24
).
Expression Quantitative Trait Loci (eQTLs)
To identify causal implications for genetic variants, eQTLs
were assessed for the top genome-wide signicant GAD-2
signals using GTEx version 7 brain tissue expression data.
Top genome-wide signicant signals on chromosomes 7 and
20 had signicant eQTLs (false discovery rate #0.05) for four
different genes: FTSJ2,RGS19,C20orf201, and OPRL1 (see
Table S7 in the online supplement). The top signals are
centered in the basal ganglia and cerebellum.
SNP-Based Heritability
SNP-based heritability using LDSC for the GAD-2 quantitative
trait was estimated to be 5.58% (SE=0.004, z-score=13.95). SNP-
based ination was mild considering the sample size and
polygenic trait studied (l=1.19); the intercept (1.026) and at-
tenuation ratio (0.1177) estimated by LDSC showed negligible
evidence for ination due to population stratication. SNP-
based heritability for the self-reported physician diagnosis of
an anxiety disorder binary trait was 8.79% (SE=0.0085, z-
score=10.34) on the liability scale assuming prevalence of 20%.
This value is similar to that reported for anxiety by Otowa et al.
(h
2
=0.095, SE=0.037, z-score=2.57) (8), depression by the PGC
(h
2
=0.087, SE=0.004, z-score=21.75) (11) and Howard et al.
(h
2
=0.089, SE=0.003, z-score=29.67) (33), and neuroticism by
Nagel et al. (h
2
=0.100, SE=0.003, z-score=33.33) (31), but some-
what lower than that reported by Meier et al. for anxiety and
stress-related disorders (h
2
=0.28, SE=0.027, z-score=10.37) (9).
Linkage Disequilibrium Score Regression Analysis
The traits most signicantly genetically correlated with GAD-
2 score were depressive symptoms (r
g
=0.81, p=1.95310
253
)
and neuroticism (r
g
=0.72, p=6.53310
253
). We also investigated
genetic correlation within the MVP cohort for GAD-2 score and
self-reported physician diagnosis of an anxiety disorder. These
were high (r
g
=0.87, p=2.39310
2119
), and higher than the phe-
notypic correlation between these traits (r=0.64, p,2.2310
216
)
(Figure 2; see also Table S5 in the online supplement).
Polygenic Risk Score (PRS) Analysis
Summary statistics from the MVP GAD-2 analysis were used
as the base data for calculating polygenic risk scores (PRSs)
(using PRSice, version 1.25 [34]). Genetic overlap between
anxiety and PTSD or major depressive disorder was tested
using the summary statistics to summary statisticspro-
cedure, using the gtx R package incorporated into PRSice, in
the PGC major depressive disorder (11) and PTSD (35)
GWASs, respectively, and overlap with case-control anxiety
disorder was tested in the largest previously available studies
(8, 9). Signicant overlap was identied: the MVP GAD-2 PRS
can explain up to 0.24% of the variance in major depressive
disorder in the PGC GWAS (p=2.05310
294
), 0.23% of the
variance in PTSD in the PGC GWAS (p=4.23310
212
), and
0.48% of the variance in both the ANGST (p=3.66310
220
)
and iPSYCH anxiety studies (p=6.68310
236
) (Table 3).
Multi-Trait-Based Conditional and Joint Analysis
Multi-trait-based conditional and joint analysis was used to
condition the GAD-2 MVP summary statistics for anxiety on
the PGC summary statistics for major depressive disorder
(11). There were no new signals and the signicance levels of
the lead ndings were reduced, but the results on chromo-
somes 3 (SATB1) and 6 (ESR1) remained genome-wide sig-
nicant. Degree of lost variance explained in the anxiety
GWAS when conditioned on major depressive disorder was
tested using LD score regression. Genetic correlation analysis
was performed between the original European American
GAD-2 GWAS summary statistics and the major depressive
disorder conditioned summary statistics, which served as an
internal control to show that the trait measured was still the
same (r
g
=1.0). Heritability dropped signicantly (p=0.021)
from 0.0558 (SE=0.0041) in the original GWAS to 0.0429
(SE=0.0038) in the conditioned GWAS.
Fine Mapping
Fine mapping in PAINTOR, version 3, was used to predict
causal SNPs using functional brain annotations (see Figure S6
in the online supplement). In one case (chromosome 6,
rs6557168) the causal SNP identied was the same as the
TABLE 2. Replication in independent cohorts in a study of genetic risk loci for anxiety
a
GAD-2 MVP
European American
Meta-Analysis
b
iPSYCH Anxiety and
Stress-Related
Disorders (9)
UK Biobank,
23andMe, and GPC
Neuroticism (31) ANGST Anxiety (8)
RSID CHR Gene p Risk Allele p Risk Allele p Risk Allele p Risk Allele
rs4603973 3 SATB1-AS1 6.18E11 G 0.181 G na na 0.0299 G
rs4390955 3 SATB1-AS1 7.78E11 A 0.851 C 5.20E04 A 0.2935 A
rs6557168 6 ESR1 1.33E09 C 0.0128 C 0.367 C 0.170 C
rs12023347 1 LINC01360 /LRRIQ3 8.88E09 T 6.61E04 T 6.85E04 T 0.00296 T
rs56226325 7 MAD1L1 2.01E08 C 6.41E04 C 6.59E08 C 0.354 C
rs6090040 20 TCEA2 3.28E08 C 0.461 A 0.444 A 0.867 C
a
Italic font indicates same direction of effect, and boldface indicates same direction of effect and nominal signicance (p=0.05). The lead SNP on chromosome
3 near the SATB1 locus, rs4603973, was not available for lookup in the UK Biobank neuroticism genome-wide association study, so we used the strongest LD proxy
available (rs4390955, R
2
=0.91, p=7.78E11). ANGST=Anxiety Neuro Genetics Study; CHR=chromosome; GAD-2=Generalized Anxiety Disorder 2-item scale;
GPC=Genetics of Personality Consortium; iPSYCH=Initiative for Integrative Psychiatric Resear ch; MVP=Million Veteran Program; RSID=reference SNP cluster ID.
b
Results from the present study.
ajp in Advance ajp.psychiatryonline.org 5
LEVEY ET AL.
GWAS lead SNP. On chromosome 20, there were several
genes in the region of our lead SNP, and several genes had
associated eQTLs. The ne mapping analysis prioritized a
likely causal SNP (rs8126001) within the 5:UTR of OPRL1.
DISCUSSION
We present the largest GWAS to date for anxiety traits,
employing a quantitative phenotype, the GAD-2 score, in
nearly 200,000 MVP sub-
jects, as well as self-reported
physician diagnosis of anxi-
ety/panic case-control phe-
notypes in .220,000 MVP
subjects. We identied novel
genetic variants in and
around several genes, some
of which have previously
known functional relation-
ships with anxiety. These
genes play roles in the
hypothalamic-pituitary-adrenal
(HPA) axis, neuronal develop-
ment, and global regulation of
gene expression.
There is high comorbidity
between anxiety, PTSD, and
depression. We used a PRS
derived from the MVP GAD-
2 analysis to identify genetic
overlap with the independent
PGC PTSD and major de-
pressive disorder GWASs
(Table3;seealsoFigureS7in
theonlinesupplement).We
found signicant genetic
overlap between these traits,
providing biological evidence
that this known clinical
comorbidity is due at least in
part to shared genetic etiology.
Additionally, we performed
multi-trait-based conditional
and joint analysis, using a prior
GWAS of depression to con-
dition the results of the present
GAD-2 GWAS. In this analysis,
we show not only that the peak
signals for anxiety are reduced
in magnitude (see Figure S3 in
the online supplement) but
also that the overall heritabil-
ityforanxietysymptomsis
diminished, from 5.58% to
4.29%, when conditioned on
genetic liability to depression.
Via linkage disequilibrium score regression, we identied
substantial genetic correlations between anxiety and numerous
other traits (Figure 2). Particularly noteworthy were positive
correlations with depression and neuroticism as well as a
negative correlation with subjective well-being (Figure 2).
These ndings replicate similar correlations found using a
case-control approach (9).
The genome-wide signicant result in African Americans
is an insertion variant that is rare outside of African ancestry
FIGURE 2. Genetic correlation between Million Veteran Program GAD-2 score in European Americans
and other traits and disorders from LD score regression in LD Hub
a
–0.6 –0.4 –0.2 0 0.2 0.4 0.6 0.8 1
Intelligence 28530673
Age of first birth 27798627
Subjective well-being 27089181
Childhood IQ 23358156
Smoking cessation 20418890
College completion 23722424
Educational attainment 25201988
Years of schooling 2016 27225129
Years of schooling 2013 23722424
Parents’ age at death 27015805
Father’s age at death 27015805
Mother’s age at death 27015805
Parkinson’s disease 19915575
HDL cholesterol 20686565
Age at menopause 26414677
Urate 23263486
Overweight 23563607
Triglycerides 20686565
Coronary artery disease 26343387
Obesity class 2 23563607
Body mass index 20935630
Rheumatoid arthritis 24390342
Obesity class 1 23563607
Waist circumference 25673412
Body fat 26833246
Waist-to-hip ratio 25673412
Fasting insulin main eect 22581228
Obesity 23563607
PGC cross-disorder analysis 23453885
Schizophrenia 25056061
Number of children ever born 27798627
Smoking initiation 20418890
Lung cancer 27488534
Squamous cell lung cancer 27488534
Lung cancer (all) 24880342
ADHD 27663945
Amyotrophic lateral sclerosis 27455348
Lung adenocarcinoma 27488534
Lung cancer (squamous cell) 24880342
Insomnia 27992416
Insomnia 28604731
Major depressive disorder 22472876
Major depressive disorder 29700475
Neuroticism 27089181
Anxiety 26754954
Neuroticism 24828478
Depressive symptoms 27089181
rg
PubMed
Measure, Trait, or Disorder Identifier
a
All plotted traits survive 0.05 false discovery rate. Full results are presented in Table S5 in the online supplement.
GAD-2=Generalized Anxiety Disorder 2-item scale; LD=linkage disequilibrium; PGC=Psychiatric Genomics
Consortium.
6ajp.psychiatryonline.org ajp in Advance
REPRODUCIBLE GENETIC RISK LOCI FOR ANXIETY
and occurs in a genomic region proposed to be under recent
selection in Europeans (36). The lead SNP is at TRPV6, which
encodes a Ca
2+
-selective membrane cation channel associ-
ated with epithelial calcium transport and homeostasis in
kidney and intestine. The lead SNP rs575403075 has an MAF
range of between 0% and 1% in non-African populations and
would fall below MAF quality control thresholds used for
common variants in most non-African populations. In indi-
viduals of African ancestry, this variant is much more com-
mon, with an MAF in our study of 5.8%. This highlights the
importance of studying genetic risks in diverse populations
otherwise these signals may be missed entirely.
The top genome-wide signicant ndings for European
Americans in the GAD-2 analysis were in and around SATB1
and the antisense gene SATB1-AS1.SATB1 is a global regu-
lator that inuences expression of multiple genes involved
in neuronal development (37). One gene modulated in
expression is Corticotropin Releasing Hormone (CRH),
encoding the protein product of the same name that plays an
essential role in the HPA axis, which has frequently been
shown to modulate stress and fear/anxiety response (38). The
CRHR1 (Corticotropin Releasing Hormone Receptor 1) gene
was genome-wide signicant in the gene-based association
analysis (p=3.60310
27
). CRHR1 has been a proposed target
for treatment for anxiety and stress-related disorders, with
evidence for anxiolytic-like effects of CRHR1 antagonists in
animal models although not yet in humans. Based on our
ndings, we speculate that individuals with differing genetic
risk that does or does not involve this pathway may differ in
their responses to CRHR1-targeted and other glucocorticoid-
targeted therapeutic agents; this may be a reasonable path-
way to address via personalized medicine, and it presents a
testable hypothesis.
The estrogen receptor ESR1 (also known as estrogen re-
ceptor alpha) has been a focus in animal models of anxiety-
like behaviors, and these have provided mechanistic validity
for the role of ESR1. Studies of estradiol administration to
ovariectomized rats and ESR1 null mice have shown con-
sistent evidence that ESR1 is involved in anxiety-like behavior
(39). Our nding of an association between ESR1 and anxiety
may have implications for our understanding of sex differ-
ences in anxiety disorders and trauma and stressor-related
disorders such as PTSD, which are more common in females
(40). Although this female predominance is partially
explained by sex-specic
exposure to certain kinds of
traumatic events (e.g., do-
mestic violence, sexual as-
sault), there may also be
differential biological con-
text provided in part by the
role of the estrogen receptor.
Our study in a predominantly
male sample identies ESR1
as genome-wide signicant.
Estrogen is important in both
sexes, and a recent review has highlighted the important role
for estrogens in men (41). Studies with larger numbers of
women will be needed to more fully investigate sex differ-
ences in genetic risk for anxiety-related traits.
Previous genetic epidemiology studies have shown that
common genetic factors can underlie anxiety and depressive
traits (42). The lead SNP from the GAD-2 GWAS near the
LINC01360 and LRRIQ3 (rs2180945) loci is nominally sig-
nicant and has the same direction of effect in the 2018 PGC
major depressive disorder analysis (p=1.434310
26
) (11). This
variant may be linked to a common risk factor for both
disorders.
One genome-wide signicant signal for GAD-2 was in a
gene-rich region on chromosome 20 near TCEA2,C20orf201,
RGS19, and OPRL1, with ne-mapping analysis prioritizing a
causal region in the latter gene. OPRLI (which encodes the
amygdala nociceptin/orphanin FQ receptor) is involved in
learning and memory and anxiety and fear-related behaviors
(43, 44) and has been hypothesized to play a role in anxiety
and stressor-related disorders such as PTSD (44). In-
terestingly, fear conditioning was also signicantly enriched
in the pathway analysis. Taken together, these observations
suggest that OPRL1 and related systems should be further
explored as targets for anxiety and stressor-related thera-
peutics. eQTL data suggest that variants in this region reg-
ulate expression of RGS19 and OPRL1 in the cerebellum and in
the basal ganglia (see Table S7 in the online supplement). The
basal ganglia have long been implicated in obsessive-
compulsive disorder and anxiety disorders. A recent re-
view discussed cerebellum-linked neurocircuitry to anxiety
and fear behaviors in rodents and in humans (45). The cer-
ebellum is thought to play an important role in anticipation/
prediction processes. Given that anxiety has been dened as
a future-oriented mood state associated with preparation for
possible, upcoming negative events(4), these results may
provide further evidence for a role for the cerebellum in fear
and anxiety.
MAD1L1 (GAD-2 lead SNP rs56226325, MAF=0.17,
p=2.01310
28
; self-reported physician diagnosis of an anxiety
disorder lead SNP rs10534613, MAF=0.41, p=4.92310
28
)is
replicated in the iPSYCH anxiety GWAS data (9) (Table2;
p=6.85310
24
) and has been associated previously with bi-
polar disorder (46). One of the lead SNPs in the iPSYCH study
is also nominally associated with anxiety in the present study
TABLE 3. Polygenic risk scores generated from the Million Veteran Program genome-wide association
study (GWAS) of GAD-2 anxiety trait and predicting into other relevant GWAS summary statistics
a
Trait (Reference Number) GWAS p Threshold SNPs Tested (N) r
2
p
PGC depression (1) 0.145 89,152 0.002445 2.05E94
PGC PTSD (35) 0.075 58,854 0.002330 4.23E12
ANGST anxiety (8) 0.415 83,033 0.004884 3.66E20
iPSYCH anxiety (9) 0.130 79,974 0.004853 6.68E36
a
ANGST5Anxiety Neuro Genetics Study; GAD-2=Generalized Anxiety Disorder 2-item scale; iPSYCH=Initiative for
Integrative Psychiatric Research; PGC=Psychiatric Genomics Consortium; PTSD=posttraumatic stress disorder. The
best-tting polygenic risk score (PRS) is shown for each trait. The rst data column contains the threshold used for the
best-tting PRS. The second column indicates the number of SNPs tested in the best-tting PRS. The r
2
is the variance
explained by the GAD-2 SNPs at the p value threshold used to create the given PRS.
ajp in Advance ajp.psychiatryonline.org 7
LEVEY ET AL.
(rs11764590, p=3.363
27
); this SNP is in LD with our lead
SNP, rs56226325 (r
2
=0.69). A recent large GWAS of bipolar
disorder identied genome-wide signicant SNPs in the
MAD1L1 locus, although their lead signal is not signicant in
our study of anxiety (rs4236274, p=0.27) (47). This locus has
also been identied among 108 genome-wide signicant loci
by the PGC schizophrenia study (rs58120505, p value=
6.43310
214
) (48), and our lead SNP is nominally signicant in
that study (rs56226325, p=1.12310
23
). This SNP is also
nominally signicant (6.71310
24
)inthe2018PGCde-
pression GWAS (11). Taken together, these observations
suggest that this locus may be a common risk factor for several
psychiatric disorders.
MAD1L1*rs56226325 is also an eQTL for expression of
FTSJ2 (seeTableS7intheonlinesupplement),andthis
variant is associated with decreased expression in the brain.
MAD1L1 is a mitochondrial RNA methyltransferase that is
important for the proper assembly of the mitochondrial
ribosome and cellular respiration (49). The protein product
of FTSJ2 is Mitochondrial rRNA Methyltransferase 2
(MRM2), which was implicated in a case study of a 7-year-
old Italian boy with a damaging mutation that reduced the
catalytic activity of MRM2, leading to an encephalopathy,
lactic acidosis, and stroke-like (MELAS) syndrome (50).
Larger-effect mutations at this locus can have devastating
effects on the brain; smaller-effect variations may be less
deleterious but still cumulatively inuence development,
which may predispose to neurological and psychiatric
disorders.
The Brainstorm Consortium has investigated shared
heritability between psychiatric and neurological disorders
(51). Consistent with their ndings, we nd very strong ge-
netic correlation between anxiety (GAD-2) and psychiatric
traits such as depression (r
g
=0.81, p=2.48310
253
) and neu-
roticism (r
g
=0.72, p=7.09310
253
) but relatively weaker ge-
netic overlap with neurological disorders. Signicant positive
genetic association is detected for amyotrophic lateral scle-
rosis (r
g
=0.39, p=3.00310
24
), and negative genetic associa-
tion with Parkinsons disease (r
g
=20.19, p=4.70310
23
), but
no genetic overlap is seen for Alzheimers disease (r
g
=0.00,
p=1.00). Further work will be needed to better discern the
implications of these ndings for understanding shared and
disparate disease mechanisms among these neuropsychiatric
conditions (11, 35, 51).
Limitations of this work include the fact that phenotypes
were based on self-reported survey data. The GAD-2 asks
questions that temporally reference the past 2 weeks.Al-
though the GAD-2 has demonstrated high sensitivity and
specicity for anxiety disorders (52), it falls short of the
desired trait (lifetime) anxiety measure. That our work re-
produces (and is reproduced by) other independent groups
who did use lifetime anxiety measures (8, 9) further supports
the utility of the GAD-2 in capturing a genetically meaningful
anxiety trait. Similarly, the question about diagnosis for
anxiety or panic that yielded our binary self-reported phy-
sician diagnosis of anxiety or panic disorder phenotype relies
on self-report. A further limitation is that MVP has pre-
dominantly male participants (92.5%). While women are
included in this analysis, clinically important interactions
between sex, phenotype, and genotype could not be
addressed. This cohort is growing, and future recruitment
will provide additional power to revisit sex-stratied analyses
of this sample. Males presumably have a higher genetic lia-
bility threshold for anxiety, as evidenced by lower rates of
anxiety disorders. Accordingly, affected males could reect
higher genetic risk than females (because they must pass a
higher threshold to be affected), which would result in
greater power to detect risk loci in a mostly male compared
with a mostly female sample.
In summary, we have identied novel variants for anxiety
by performing GWASs in the large MVP cohort. We repli-
cated results in our GWASs for top ndings from recent
anxiety and other relevant anxiety-related GWASs. We also
identied signicant genetic overlap with major depressive
disorder, PTSD, and neuroticism using polygenic risk scores
and LD score regression. This work provides additional ge-
netic evidence for the overlap between disorders that are
frequently comorbid with anxiety and presents new mo-
lecular targets for investigation with a longer view toward the
development of new treatments.
AUTHOR AND ARTICLE INFORMATION
Division of Human Genetics, Department of Psychiatry, Yale University
School of Medicine, New Haven, Conn., and Department of Psychiatry,
Veterans Affairs Connecticut Healthcare Center, West Haven, Conn.
(Levey, Gelernter, Polimanti, Zhou, Cheng); VA Clinical Epidemiology
Research Center, VA Connecticut Healthcare System, West Haven, Conn.
(Aslan, Concato, Radhakrishnan); Department of Medicine, Yale University
School of Medicine, New Haven, Conn. (Aslan, Concato); Massachusetts
Veterans Epidemiology Research and Information Center, VA Boston
Healthcare System, Boston (Quaden); College of Medicine, University of
Kentucky, Lexington (Radhakrishnan); Department of Medical Epidemi-
ology and Biostatistics, Karolinska Institutet, Stockholm (Bryois, Sullivan);
Departments of Genetics and Psychiatry, University of North Carolina,
Chapel Hill (Sullivan); Psychiatry Service, VA San Diego Healthcare System,
San Diego, and Departments of Psychiatry and of Family Medicine and
Public Health, University of California San Diego, La Jolla (Stein).
Send correspondence to Dr. Gelernter (joel.gelernter@yale.edu) and
Dr. Stein (mstein@health.ucsd.edu).
Drs. Levey, Gelernter, and Stein contributed equally to this work.
Supported by funding from the Veterans Affairs Ofce of Research and
Development Million Veteran Program grant MVP011 and VA Cooperative
Studies Program CSP575B.
The authors thank the veterans who participate in the Million Veteran
Program.
Dr. Sullivan has served on an advisory committee and received grant
support from Lundbeck and served on a scientic advisory board for Pzer.
Dr. Gelernter is named as co-inventor on PCT patent application 15/
878,640 (genotype-guided dosing of opioid agonists), led January 24,
2018. Dr. Stein has served as a consultant for Aptinyx, Bionomics, Janssen,
Jazz Pharmaceuticals, Neurocrine, Pzer, and Oxeia Biopharmaceuticals.
The other authors report no nancial relationships with commercial
interests.
Received March 8, 2019; revisions received August 9 and October 14,
2019; accepted October 15, 2019.
8ajp.psychiatryonline.org ajp in Advance
REPRODUCIBLE GENETIC RISK LOCI FOR ANXIETY
REFERENCES
1. GBD 2017 Disease and Injury Incidence and Prevalence Collabo-
rators: Global, regional, and national incidence, prevalence, and
years lived with disability for 354 diseases and injuries for
195 countries and territories, 19902017: a systematic analysis for the
Global Burden of Disease Study 2017. Lancet 2018; 392:17891858
2. Roehrig C: Mental disorders top the list of the most costly conditions
in the United States: $201 billion. Health Aff (Millwood) 2016; 35:
11301135
3. Stein MB, Craske MG: Treating Anxiety in 2017: optimizing care to
improve outcomes. JAMA 2017; 318:235236
4. Craske MG, Rauch SL, Ursano R, et al: What is an anxiety disorder?
Depress Anxiety 2009; 26:10661085
5. American Psychiatric Association: Diagnostic and Statistical Manual
of Mental Disorders, 5th ed (DSM-5). Washington, DC, American
Psychiatric Association, 2013
6. Craske MG, Stein MB, Eley TC, et al: Anxiety disorders. Nat Rev Dis
Primers 2017; 3:17024
7. Zinbarg RE, Mineka S, Bobova L, et al: Testing a hierarchical model
of neuroticism and its cognitive facets: latent structure and pro-
spective prediction of rst onsets of anxiety and unipolar mood
disorders during 3 years in late adolescence. Clin Psychol Sci 2016; 4:
805824
8. Otowa T, Hek K, Lee M, et al: Meta-analysis of genome-wide as-
sociation studies of anxiety disorders. Mol Psychiatry 2016; 21:
13911399
9. Meier SM, Trontti K, Purves KL, et al: Genetic variants associated
with anxiety and stress-related disorders: a genome-wide associa-
tion study and mouse-model study. JAMA Psychiatry (Epub ahead
of print, May 22, 2019)
10. Purves KL, Coleman JR, Rayner C, et al: The common genetic ar-
chitecture of anxiety disorders. bioRxiv, October 16, 2017 (https://
doi.org/10.1101/203844)
11. Wray NR, Ripke S, Mattheisen M, et al: Genome-wide association
analyses identify 44 risk variants and rene the genetic architecture
of major depression. Nat Genet 2018; 50:668681
12. Baxter AJ, Vos T, Scott KM, et al: The global burden of anxiety
disorders in 2010. Psychol Med 2014; 44:23632374
13. Gelernter CS, Uhde TW, Cimbolic P, et al: Cognitive-behavioral and
pharmacological treatments of social phobia: a controlled study.
Arch Gen Psychiatry 1991; 48:938945
14. Stein MB, Sareen J: Anxiety disorders, in American Psychiatric
Association Publishing Textbook of Psychiatry, 7th ed. Edited by
Roberts LW. Washington, DC, American Psychiatric Association
Publishing, 2018, pp 341370
15. Loerinc AG, Meuret AE, Twohig MP, et al: Response rates for CBT
for anxiety disorders: need for standardized criteria. Clin Psychol
Rev 2015; 42:7282
16. Lee JK, Wang J, Sa JK, et al: Spatiotemporal genomic architecture
informs precision oncology in glioblastoma. Nat Genet 2017; 49:
594599
17. Stein MB, Smoller JW: Precision psychiatry: will genomic medicine
lead the way? JAMA Psychiatry 2018; 75:663664
18. Boyle EA, Li YI, Pritchard JK: An expanded view of complex traits:
from polygenic to omnigenic. Cell 2017; 169:11771186
19. Sugrue LP, Desikan RS: What are polygenic scores and why are they
important? JAMA 2019; 321:18201821
20. Gaziano JM, Concato J, Brophy M, et al: Million Veteran Program: a
mega-biobank to study genetic inuences on health and disease.
J Clin Epidemiol 2016; 70:214223
21. Gelernter J, Sun N, Polimanti R, et al: Genome-wide association
study of post-traumatic stress disorder reexperiencing symptoms
in .165,000 US veterans. Nat Neurosci 2019; 22:13941401
22. Gelernter J, Sun N, Polimanti R, et al: Genome-wide association
study of maximum habitual alcohol intake in .140,000 U.S. Euro-
pean and African American veterans yields novel risk loci. Biol
Psychiatry 2019; 86:365376
23. Kroenke K, Spitzer RL, Williams JB, et al: The Patient Health
Questionnaire Somatic, Anxiety, and Depressive Symptom Scales: a
systematic review. Gen Hosp Psychiatry 2010; 32:345359
24. Purcell S, Neale B, Todd-Brown K, et al: PLINK: a tool set for whole-
genome association and population-based linkage analyses. Am J
Hum Genet 2007; 81:559575
25. Price AL, Patterson NJ, Plenge RM, et al: Principal components
analysis corrects for stratication in genome-wide association
studies. Nat Genet 2006; 38:904909
26. Zheng J, Erzurumluoglu AM, Elsworth BL, et al: LD Hub: a cen-
tralized database and web interface to perform LD score regression
that maximizes the potential of summary level GWAS data for SNP
heritability and genetic correlation analysis. Bioinformatics 2017; 33:
272279
27. Zhu Z, Zheng Z, Zhang F, et al: Causal associations between risk
factors and common diseases inferred from GWAS summary data.
Nat Commun 2018; 9:224
28. Watanabe K, Taskesen E, van Bochoven A, et al: Functional mapping
and annotation of genetic associations with FUMA. Nat Commun
2017; 8:1826
29. Kichaev G, Yang WY, Lindstrom S, et al: Integrating functional data
to prioritize causal variants in statistical ne-mapping studies. PLoS
Genet 2014; 10:e1004722
30. Maurano MT, Humbert R, Rynes E, et al: Systematic localization of
common disease-associated variation in regulatory DNA. Science
2012; 337:11901195
31. Nagel M, Jansen PR, Stringer S, et al: Meta-analysis of genome-wide
association studies for neuroticism in 449,484 individuals identies
novel genetic loci and pathways. Nat Genet 2018; 50:920927
32. Krämer A, Green J, Pollard J Jr, et al: Causal analysis approaches in
Ingenuity Pathway Analysis. Bioinformatics 2014; 30:523530
33. Howard DM, Adams MJ, Clarke TK, et al: Genome-wide meta-
analysis of depression identies 102 independent variants and
highlights the importance of the prefrontal brain regions. Nat
Neurosci 2019; 22:343352
34. Euesden J, Lewis CM, OReilly PF: PRSice: Polygenic Risk Score
software. Bioinformatics 2015; 31:14661468
35. Duncan LE, Ratanatharathorn A, Aiello AE, et al: Largest GWAS of
PTSD (N=20 070) yields genetic overlap with schizophrenia and sex
differences in heritability. Mol Psychiatry 2018; 23:666673
36. Akey JM, Eberle MA, Rieder MJ, et al: Population history and natural
selection shape patterns of genetic variation in 132 genes. PLoS Biol
2004; 2:e286
37. Balamotis MA, Tamberg N, Woo YJ, et al: Satb1 ablation alters
temporal expression of immediate early genes and reduces dendritic
spine density during postnatal brain development. Mol Cell Biol
2012; 32:333347
38. Zorrilla EP, Valdez GR, Nozulak J, et al: Effects of antalarmin, a CRF
type 1 receptor antagonist, on anxiety-like behavior and motor ac-
tivation in the rat. Brain Res 2002; 952:188199
39. Borrow AP, Handa RJ: Estrogen receptors modulation of anxiety-
like behavior. Vitam Horm 2017; 103:2752
40. Haskell SG, Gordon KS, Mattocks K, et al: Gender differences in rates
of depression, PTSD, pain, obesity,and military sexual trauma among
Connecticut war veterans of Iraq and Afghanistan. J Womens Health
(Larchmt) 2010; 19:267271
41. Cooke PS, Nanjappa MK, Ko C, et al: Estrogens in male physiology.
Physiol Rev 2017; 97:9951043
42. Mosing MA, Gordon SD, Medland SE, et al: Genetic and environ-
mental inuences on the co-morbidity between depression, panic
disorder, agoraphobia, and social phobia: a twin study. Depress
Anxiety 2009; 26:10041011
43. Andero R: Nociceptin and the nociceptin receptor in learning and
memory.P rog Neuropsychopharmacol Biol Psychiatry 2015; 62:4550
44. Rekik K, Faria Da Silva R, Colom M, et al: Activation of nociceptin/
orphanin FQ receptors inhibits contextual fear memory reconsoli-
dation. Neuropharmacology 2017; 125:3949
ajp in Advance ajp.psychiatryonline.org 9
LEVEY ET AL.
45. Moreno-Rius J: The cerebellum in fear and anxiety-related dis-
orders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 85:
2332
46. Cichon S, Mühleisen TW, Degenhardt FA, et al: Genome-wide as-
sociation study identies genetic variation in neurocan as a sus-
ceptibility factor for bipolar disorder. Am J Hum Genet 2011; 88:
372381
47. Hou L, Bergen SE, Akula N, et al: Genome-wide association study of
40,000 individuals identies two novel loci associated with bipolar
disorder. Hum Mol Genet 2016; 25:33833394
48. Ripke S, Neale BM, Corvin A, et al: Biological insights from
108 schizophrenia-associated genetic loci. Nature 2014; 511:421427
49. Rorbach J, Boesch P, Gammage PA, et al: MRM2 and MRM3 are
involved in biogenesis of the large subunit of the mitochondrial
ribosome. Mol Biol Cell 2014; 25:25422555
50. Garone C, DSouza AR, Dallabona C, et al: Defective mitochondrial
rRNA methyltransferase MRM2 causes MELAS-like clinical syn-
drome. Hum Mol Genet 2017; 26:42574266
51. Anttila V, Bulik-Sullivan B, Finucane HK, et al: Analysis of shared
heritability in common disorders of the brain. Science 2018; 360:
eaap8757
52. Kroenke K, Spitzer RL, Williams JB, et al: Anxiety disorders in
primary care: prevalence, impairment, comorbidity, and detection.
Ann Intern Med 2007; 146:317325
10 ajp.psychiatryonline.org ajp in Advance
REPRODUCIBLE GENETIC RISK LOCI FOR ANXIETY
... Previous genome-wide association study (GWAS) meta-analyses failed to identify specific genetic loci for panic disorder due to limited sample sizes 7 . In addition, anxiety GWAS research has often grouped panic disorder with other anxiety disorders 8,9 . It is critical to examine panic attacks and disorder independently and at scale because their episodic nature, distinct symptomatology, and emphasis on fear may reflect distinct genetic aetiology compared to other anxiety disorders that are characterized by persistent worry. ...
Preprint
Introductory paragraph Panic attacks, sudden episodes of intense fear accompanied by physical and psychological symptoms, affect approximately 23% of the population 1,2 . Panic disorder, diagnosed in 2– 4% ³ , involves recurrent attacks followed by persistent worry about further attacks, leading to functional impairment and avoidance behaviours 1,2 . We conducted genome-wide association meta-analyses of panic attacks and panic disorder (61,746 and 29,775 cases, respectively, and 105,814 controls), identifying the first genome-wide significant variants for both traits (16 for panic attacks; 7 for panic disorder). Geneset analysis using single-cell RNA sequencing data implicated afferent neurons from the eye, heart, and lung in panic attacks, suggesting a critical role for sensory processing and interoceptive awareness. Further analyses suggested that these associations generalize to other psychiatric disorders. These findings offer novel insights into the biological mechanisms underlying panic, the role of afferent neurons, and may inform the development of more targeted and effective interventions.
... Extending prior research, 32,35,48,49 our findings demonstrate shared genetic risk between clinical diagnoses and corresponding symptoms in the general population. This is aligned with dimensional frameworks such as RDoC 50 and HiTOP 51 , which conceptualize All rights reserved. ...
Preprint
Importance: Polygenic scores are increasingly used to estimate genetic liability for psychiatric disorders. However, their limited specificity to the disorders for which they are derived limits their clinical and research utility. Objective: To determine whether associations between polygenic scores for major psychiatric disorders and psychopathology outcomes are primarily driven by transdiagnostic (p) or disorder-specific genetic risk. Design: Cross-sectional study. Analyses were conducted from May 2024 to October 2024. Setting: Population-based sample from the Twins Early Development Study. Participants: Twins born in England and Wales between 1994 and 1996. Mental health data were collected from 2021 to 2023 when participants were aged 25 to 28 years. Participants with available genetic data and at least one quantitative symptom score included. Main outcomes and measures: Quantitative symptom scores and self-reported psychiatric diagnoses. Associations were tested using generalized estimating equations for three types of polygenic scores: uncorrected polygenic scores; a transdiagnostic polygenic score indexing shared genetic variance across 11 psychiatric conditions (p); and residual disorder-specific scores corrected for p (non-p). Results: Analyses included between 5,789 and 6,546 (mean [SD] age, 26.4 [0.93] years; 7,244 [51.6%] female). The polygenic score for p consistently showed stronger associations with symptom scores than uncorrected polygenic scores, while associations with self-reported diagnoses were similar. Most uncorrected polygenic scores exhibited extensive cross-trait associations, which were substantially attenuated accounting for p, suggesting that much of the genetic signal captured by polygenic scores reflects transdiagnostic liability. Some non-p polygenic scores retained associations with their corresponding-traits, indicating residual specificity. Conclusions and relevance: In this population-based sample of young adults, associations between polygenic scores and psychopathology outcomes primarily reflect transdiagnostic genetic risk, with limited evidence of disorder-specificity. Accounting for transdiagnostic genetic liability could improve the specificity and interpretability of polygenic scores.
... Anxiety disorders are also highly comorbid with other mental health disorders including alcohol use disorder (AUD), with individuals diagnosed with AUD having 2.1 times greater risk of having any anxiety disorder compared to non-alcohol users (OR 2.1, 95% CI 2.0-2.2) [7]. Numerous human GWAS studies have identified loci associated with anxiety disorders, implicating genes including CTNND1, RAB27B [8], SATB1, MAD1L1 [9], and others. However, the individual genes associated with anxiety disorders are thought to be of small effect [10,11], with recent estimates indicating as many as 12,900 potential risk loci [12]. ...
Preprint
Anxiety disorders are the most common class of psychiatric disorder. Risk for anxiety disorders is thought to be influenced by many genes, each contributing a small effect. The light-dark box behavioral assay was designed to measure anxiety-like behavior in rodents. Diversity Outbred (DO) mice were designed for high-resolution quantitative trait loci (QTL) mapping on a genetically-diverse background. Here, we studied a population of 518 male DO mice for anxiety-like and locomotor behaviors from a light-dark box assay. Multivariate analysis of behavioral data identified two major subgroups of animals differing in basal anxiety behavior and subsequent ethanol consumption patterns. Behavioral QTL analysis identified a significant locus on Chromosome 14 associated with 3 anxiety-like behavioral phenotypes. Haplotype analysis revealed an effect of C57BL/6J alleles at this locus, with mice carrying those alleles exhibiting more anxiety-like behavior. An additional 9 suggestive loci were identified. Genes located within the confidence intervals for the Chromosome 14 locus were analyzed for coding sequence polymorphisms, prefrontal cortex expression QTLs, human GWAS data, and additional data sets related to psychiatric conditions including substance use. Results prioritized two candidate genes, Tbc1d4 and Lmo7, for further study. These results represent the highest-resolution genetic mapping of light-dark box behaviors in mice to date, revealing insights into the complex biology of anxiety disorders. Additionally the studies identify striking subgroups of animals where basal anxiety-like behavior predicts eventual ethanol consumption phenotypes.
... ADs have multiple factors that influence their development, among which we find genetic factors, as some genome-wide association studies have pointed out some singlenucleotide polymorphisms (SNPs) on genes, such as STAB1, ESR1, and MAD1L1, among others [29]. ...
Article
Full-text available
Anxiety is a fear response that triggers a stress reaction with the purpose of defending against or avoiding danger, which is considered physiological, until it becomes excessive and persistent, affecting daily life activities. Non-coding RNAs have been explored in terms of their relationship with diseases, and several of them, such as miRNAs, have been found to be key factors in the development of diseases. Specifically, the expression of long non-coding RNAs (lncRNAs) has been implicated in the development of anxiety through various mechanisms such as nervous system development, synaptic function, neurotransmitter regulation, and neuroinflammation. However, several recent reviews have explored the roles of lncRNAs in various mental diseases (mainly in schizophrenia), and considering that existing reviews do not cover the interaction between lncRNAs and aspects such as neuroimmunity in anxiety disorder pathophysiology, the aim of this literature review is to summarize the current knowledge about the contributions of lncRNAs to the molecular and cellular mechanisms underlying the pathogenesis of anxiety disorders. Additionally, we explore their potential applications in the diagnosis, as well as possible treatment approaches, of these disorders, challenges, and current limitations.
... In particular, in females, regions whose volumetric changes were associated with depressive-and anxiety-like behaviors highly express genes previously implicated in the process of protein localization to the cell surface. The top correlated genes associated with this phenotype suggest that alterations observed in this LV may be attributed to altered synaptic connectivity [Gtdc1, Slm2, Kcnj6, Nos1ap] (11,(50)(51)(52)(53)(54)69) in regions involved in the response of the HPA axis [Satb1] (70,71), which may underlie alterations in depressive-and anxiety-like behavior, and possibly somatic symptoms like sleep disturbances [Chrm3] (72). Overall our findings suggest that the alterations captured by structural MRI are attributed to multiple biological mechanisms and generate new hypotheses about behavioral domains that can be affected by chronic stress. ...
Preprint
Full-text available
Background: Stress is a significant risk factor for depression and anxiety, two highly comorbid disorders with sex differences in symptom presentation and prevalence. The Chronic Variable Stress (CVS) mouse model is a useful method for examining sex-specific susceptibility, as stress can be titrated in a sex-specific manner to produce depressive- and anxiety-like behaviours across both sexes. However, the sex-specific mechanisms regarding how CVS reorganizes brain anatomy remain unclear. Methods: Using structural magnetic resonance imaging (MRI), we provide the first whole-brain characterization of neuroanatomical changes induced by 6 or 28 days of exposure to CVS in female and male mice, respectively, and their association to behavior. We then examined the structural connectome underlying sex-specific latent dimensions of stress-susceptibility and potential molecular mechanisms using spatial gene expression analyses. Results: CVS induced significant neuroanatomical changes in regions already implicated in depression in both sexes (e.g. nucleus accumbens and hippocampus) as well as female- and male-specific neuroanatomical changes. In females, these changes were associated with both depressive- and anxiety-like behavior. While in males, we identified two orthogonal dimensions of neuroanatomical changes associated with anxiety-like behavior or social preference. These latent dimensions are associated with sex-specific hub regions and, in females, were associated with genes enriched for protein localization to the cell surface. Conclusion: Our findings indicate that different durations of CVS result in similar neuroanatomical changes in both sexes, however the direction of change and association to behavior is sex-specific. In females, these changes may be attributed to alterations in synaptic connectivity.
... 6 Mood and anxiety disorders tend to co-occur, 7 respond to similar pharmacological and psychological treatments 8,9 and have been indicated to involve similar neurobiological mechanisms. 10 Despite extensive research, their aetiology is still incompletely understood; however, it is evident that genetic predisposition plays a role, with common [11][12][13][14] as well as rare variants 15,16 having been associated with both disorders. There is abundant evidence that the two disorders share genetic liability. ...
Article
Full-text available
Background Mood and anxiety disorders co-occur and share symptoms, treatments and genetic risk, but it is unclear whether combining them into a single phenotype would better capture genetic variation. The contribution of common genetic variation to these disorders has been investigated using a range of measures; however, the differences in their ability to capture variation remain unclear, while the impact of rare variation is mostly unexplored. Aims We aimed to explore the contributions of common genetic variation and copy number variations associated with risk of psychiatric morbidity (P-CNVs) to different measures of internalising disorders. Method We investigated eight definitions of mood and anxiety disorder, and a combined internalising disorder, derived from self-report questionnaires, diagnostic assessments and electronic healthcare records (EHRs). Association of these definitions with polygenic risk scores (PRSs) of major depressive disorder and anxiety disorder, as well as presence of a P-CNV, was assessed. Results The effect sizes of both PRSs and P-CNVs were similar for mood and anxiety disorder. Compared to mood and anxiety disorder, internalising disorder resulted in higher prediction accuracy for PRSs, and increased significance of associations with P-CNVs for most definitions. Comparison across the eight definitions showed that PRSs had higher prediction accuracy and effect sizes for stricter definitions, whereas P-CNVs were more strongly associated with EHR- and self-report-based definitions. Conclusions Future studies may benefit from using a combined internalising disorder phenotype, and may need to consider that different phenotype definitions may be more informative depending on whether common or rare variation is studied.
... panic disorder, agoraphobia, generalized anxiety disorder [GAD], specific phobia, and social phobia) and subclinical anxiety (i.e. GAD-2 scores), we combined three GWAS (Levey et al., 2020;Otowa et al., 2016;Purves et al., 2020) via multi-trait analysis of GWAS (Turley et al., 2018). ...
Article
Full-text available
Background There is considerable comorbidity between externalizing (EXT) and internalizing (INT) psychopathology. Understanding the shared genetic underpinnings of these spectra is crucial for advancing knowledge of their biological bases and informing empirical models like the Research Domain Criteria (RDoC) and Hierarchical Taxonomy of Psychopathology (HiTOP). Methods We applied genomic structural equation modeling to summary statistics from 16 EXT and INT traits in individuals genetically similar to European reference panels (EUR-like; n = 16,400 to 1,074,629). Traits included clinical (e.g. major depressive disorder, alcohol use disorder) and subclinical measures (e.g. risk tolerance, irritability). We tested five confirmatory factor models to identify the best fitting and most parsimonious genetic architecture and then conducted multivariate genome-wide association studies (GWAS) of the resulting latent factors. Results A two-factor correlated model, representing EXT and INT spectra, provided the best fit to the data. There was a moderate genetic correlation between EXT and INT (r = 0.37, SE = 0.02), with bivariate causal mixture models showing extensive overlap in causal variants across the two spectra (94.64%, SE = 3.27). Multivariate GWAS identified 409 lead genetic variants for EXT, 85 for INT, and 256 for the shared traits. Conclusions The shared genetic liabilities for EXT and INT identified here help to characterize the genetic architecture underlying these frequently comorbid forms of psychopathology. The findings provide a framework for future research aimed at understanding the shared and distinct biological mechanisms underlying psychopathology, which will help to refine psychiatric classification systems and potentially inform treatment approaches.
Article
Full-text available
Increasing prevalence of cannabis use and cannabis use disorder (CanUD) may increase risk for psychiatric disorders. We evaluated the relationships between these cannabis traits and a range of psychiatric traits, running global and local genetic correlations, genomic structural equation modeling, colocalization analyses and Mendelian randomization analyses for causality. Global genetic analyses identified significantly different correlations between CanUD and cannabis use. A variant in strong linkage disequilibrium to one regulating CHRNA2 was significantly shared by CanUD and schizophrenia in colocalization analysis and included in a significant region in local genetic correlations between these traits. A three-factor model from genomic structural equation modeling showed that CanUD and cannabis use partially map together onto a factor with major depressive disorder and ADHD. In terms of causality, CanUD showed bidirectional causal relationships with most tested psychiatric disorders, differently from cannabis use. Increasing use of cannabis can increase rates of psychiatric disorders over time, especially in individuals who progress from cannabis use to CanUD.
Article
Full-text available
Background Innovation in psychiatric therapeutics has stagnated on known mechanisms. Psychiatric genome-wide association studies (GWAS) have identified hundreds of genome-wide significant (GWS) loci that have rapidly advanced our understanding of disease etiology. However, whether these results can be leveraged to improve clinical treatment for specific psychiatric disorders remains poorly understood. Methods In this proof-of-principal evaluation of GWAS clinical utility, we test whether the targets of drugs used to treat Attention Deficit Hyperactivity Disorder (ADHD), Bipolar Disorder (BiP), Generalized Anxiety Disorder (GAD), Major Depressive Disorder (MDD), Post-Traumatic Stress Disorder (PTSD), Schizophrenia (SCZ), Substance Use Disorders (SUDs), and insomnia (INS), are enriched for GWAS meta-analysis findings. Results The genes coding for treatment targets of medications used to SCZ, BiP, MDD, and SUDs (but not ADHD, PTSD, GAD, or INSOM) are enriched for GWS loci identified in their respective GWAS (ORs: 2.78-27.63; all ps <1.15e-3). Enrichment is largely driven by the presence of a GWS locus or loci within a gene coding for a drug target (i.e., proximity matching). Broadly, additional annotation (i.e., functional: Combined Annotation Dependent Depletion [CADD] scores, regulomeDB scores, eQTL, chromatin loop, and gene region; statistical: effect size of genome-wide significant SNPs; Z-score of SNPs; number of drug targets implicated by GWAS), with the exception of weighting by the largest SNP effect size, does not further improve enrichment across disorders. Evaluation of prior smaller GWAS reveal that more recent larger GWAS improve enrichment. Conclusions GWAS results may assist in the prioritization of medications for future psychopharmaceutical research.
Preprint
Full-text available
Anxiety disorders are common, complex psychiatric disorders with twin heritabilities of 30-60%. We conducted a genome-wide association study of Lifetime Anxiety Disorder (n = 83 565) and an additional Current Anxiety Symptoms (n= 77 125) analysis. The liability scale common variant heritability estimate for Lifetime Anxiety Disorder was 26%, and for Current Anxiety Symptoms was 31%. Five novel genome-wide significant loci were identified including an intergenic region on chromosome 9 that has previously been associated with neuroticism, and a locus overlapping the BDNF receptor gene, NTRK2 . Anxiety showed significant genetic correlations with depression and insomnia as well as coronary artery disease, mirroring findings from epidemiological studies. We conclude that common genetic variation accounts for a substantive proportion of the genetic architecture underlying anxiety.
Article
Full-text available
Post-traumatic stress disorder (PTSD) is a major problem among military veterans and civilians alike, yet its pathophysiology remains poorly understood. We performed a genome-wide association study and bioinformatic analyses, which included 146,660 European Americans and 19,983 African Americans in the US Million Veteran Program, to identify genetic risk factors relevant to intrusive reexperiencing of trauma, which is the most characteristic symptom cluster of PTSD. In European Americans, eight distinct significant regions were identified. Three regions had values of P < 5 × 10⁻¹⁰: CAMKV; chromosome 17 closest to KANSL1, but within a large high linkage disequilibrium region that also includes CRHR1; and TCF4. Associations were enriched with respect to the transcriptomic profiles of striatal medium spiny neurons. No significant associations were observed in the African American cohort of the sample. Results in European Americans were replicated in the UK Biobank data. These results provide new insights into the biology of PTSD in a well-powered genome-wide association study.
Article
Full-text available
Major depression is a debilitating psychiatric illness that is typically associated with low mood and anhedonia. Depression has a heritable component that has remained difficult to elucidate with current sample sizes due to the polygenic nature of the disorder. To maximize sample size, we meta-analyzed data on 807,553 individuals (246,363 cases and 561,190 controls) from the three largest genome-wide association studies of depression. We identified 102 independent variants, 269 genes, and 15 genesets associated with depression, including both genes and gene pathways associated with synaptic structure and neurotransmission. An enrichment analysis provided further evidence of the importance of prefrontal brain regions. In an independent replication sample of 1,306,354 individuals (414,055 cases and 892,299 controls), 87 of the 102 associated variants were significant after multiple testing correction. These findings advance our understanding of the complex genetic architecture of depression and provide several future avenues for understanding etiology and developing new treatment approaches. © 2019, The Author(s), under exclusive licence to Springer Nature America, Inc.
Article
Full-text available
Background The Global Burden of Diseases, Injuries, and Risk Factors Study 2017 (GBD 2017) includes a comprehensive assessment of incidence, prevalence, and years lived with disability (YLDs) for 354 causes in 195 countries and territories from 1990 to 2017. Previous GBD studies have shown how the decline of mortality rates from 1990 to 2016 has led to an increase in life expectancy, an ageing global population, and an expansion of the non-fatal burden of disease and injury. These studies have also shown how a substantial portion of the world's population experiences non-fatal health loss with considerable heterogeneity among different causes, locations, ages, and sexes. Ongoing objectives of the GBD study include increasing the level of estimation detail, improving analytical strategies, and increasing the amount of high-quality data. Methods We estimated incidence and prevalence for 354 diseases and injuries and 3484 sequelae. We used an updated and extensive body of literature studies, survey data, surveillance data, inpatient admission records, outpatient visit records, and health insurance claims, and additionally used results from cause of death models to inform estimates using a total of 68 781 data sources. Newly available clinical data from India, Iran, Japan, Jordan, Nepal, China, Brazil, Norway, and Italy were incorporated, as well as updated claims data from the USA and new claims data from Taiwan (province of China) and Singapore. We used DisMod-MR 2.1, a Bayesian meta-regression tool, as the main method of estimation, ensuring consistency between rates of incidence, prevalence, remission, and cause of death for each condition. YLDs were estimated as the product of a prevalence estimate and a disability weight for health states of each mutually exclusive sequela, adjusted for comorbidity. We updated the Socio-demographic Index (SDI), a summary development indicator of income per capita, years of schooling, and total fertility rate. Additionally, we calculated differences between male and female YLDs to identify divergent trends across sexes. GBD 2017 complies with the Guidelines for Accurate and Transparent Health Estimates Reporting. Findings Globally, for females, the causes with the greatest age-standardised prevalence were oral disorders, headache disorders, and haemoglobinopathies and haemolytic anaemias in both 1990 and 2017. For males, the causes with the greatest age-standardised prevalence were oral disorders, headache disorders, and tuberculosis including latent tuberculosis infection in both 1990 and 2017. In terms of YLDs, low back pain, headache disorders, and dietary iron deficiency were the leading Level 3 causes of YLD counts in 1990, whereas low back pain, headache disorders, and depressive disorders were the leading causes in 2017 for both sexes combined. All-cause age-standardised YLD rates decreased by 3·9% (95% uncertainty interval [UI] 3·1–4·6) from 1990 to 2017; however, the all-age YLD rate increased by 7·2% (6·0–8·4) while the total sum of global YLDs increased from 562 million (421–723) to 853 million (642–1100). The increases for males and females were similar, with increases in all-age YLD rates of 7·9% (6·6–9·2) for males and 6·5% (5·4–7·7) for females. We found significant differences between males and females in terms of age-standardised prevalence estimates for multiple causes. The causes with the greatest relative differences between sexes in 2017 included substance use disorders (3018 cases [95% UI 2782–3252] per 100 000 in males vs s1400 [1279–1524] per 100 000 in females), transport injuries (3322 [3082–3583] vs 2336 [2154–2535]), and self-harm and interpersonal violence (3265 [2943–3630] vs 5643 [5057–6302]). Interpretation Global all-cause age-standardised YLD rates have improved only slightly over a period spanning nearly three decades. However, the magnitude of the non-fatal disease burden has expanded globally, with increasing numbers of people who have a wide spectrum of conditions. A subset of conditions has remained globally pervasive since 1990, whereas other conditions have displayed more dynamic trends, with different ages, sexes, and geographies across the globe experiencing varying burdens and trends of health loss. This study emphasises how global improvements in premature mortality for select conditions have led to older populations with complex and potentially expensive diseases, yet also highlights global achievements in certain domains of disease and injury.
Article
Full-text available
Disorders of the brain can exhibit considerable epidemiological comorbidity and often share symptoms, provoking debate about their etiologic overlap. We quantified the genetic sharing of 25 brain disorders from genome-wide association studies of 265,218 patients and 784,643 control participants and assessed their relationship to 17 phenotypes from 1,191,588 individuals. Psychiatric disorders share common variant risk, whereas neurological disorders appear more distinct from one another and from the psychiatric disorders. We also identified significant sharing between disorders and a number of brain phenotypes, including cognitive measures. Further, we conducted simulations to explore how statistical power, diagnostic misclassification, and phenotypic heterogeneity affect genetic correlations. These results highlight the importance of common genetic variation as a risk factor for brain disorders and the value of heritability-based methods in understanding their etiology.
Article
Full-text available
Neuroticism is an important risk factor for psychiatric traits, including depression 1 , anxiety2,3, and schizophrenia4-6. At the time of analysis, previous genome-wide association studies7-12 (GWAS) reported 16 genomic loci associated to neuroticism10-12. Here we conducted a large GWAS meta-analysis (n = 449,484) of neuroticism and identified 136 independent genome-wide significant loci (124 new at the time of analysis), which implicate 599 genes. Functional follow-up analyses showed enrichment in several brain regions and involvement of specific cell types, including dopaminergic neuroblasts (P = 3.49 × 10-8), medium spiny neurons (P = 4.23 × 10-8), and serotonergic neurons (P = 1.37 × 10-7). Gene set analyses implicated three specific pathways: neurogenesis (P = 4.43 × 10-9), behavioral response to cocaine processes (P = 1.84 × 10-7), and axon part (P = 5.26 × 10-8). We show that neuroticism's genetic signal partly originates in two genetically distinguishable subclusters 13 ('depressed affect' and 'worry'), suggesting distinct causal mechanisms for subtypes of individuals. Mendelian randomization analysis showed unidirectional and bidirectional effects between neuroticism and multiple psychiatric traits. These results enhance neurobiological understanding of neuroticism and provide specific leads for functional follow-up experiments.
Article
Neuroticism is an important risk factor for psychiatric traits, including depression 1, anxiety 2,3, and schizophrenia 4-6 . At the time of analysis, previous genome-wide association studies 7-12 (GWAS) reported 16 genomic loci associated to neuroticism 10-12 . Here we conducted a large GWAS meta-analysis (n = 449,484) of neuroticism and identified 136 independent genome-wide significant loci (124 new at the time of analysis), which implicate 599 genes. Functional follow-up analyses showed enrichment in several brain regions and involvement of specific cell types, including dopaminergic neuroblasts (P = 3.49 × 10-8), medium spiny neurons (P = 4.23 × 10-8), and serotonergic neurons (P = 1.37 × 10-7). Gene set analyses implicated three specific pathways: neurogenesis (P = 4.43 × 10-9), behavioral response to cocaine processes (P = 1.84 × 10-7), and axon part (P = 5.26 × 10-8). We show that neuroticism's genetic signal partly originates in two genetically distinguishable subclusters 13 ('depressed affect' and 'worry'), suggesting distinct causal mechanisms for subtypes of individuals. Mendelian randomization analysis showed unidirectional and bidirectional effects between neuroticism and multiple psychiatric traits. These results enhance neurobiological understanding of neuroticism and provide specific leads for functional follow-up experiments.
Article
Mendelian disorders and monogenic traits result from combinations of variants in 1 or a few genes that have a large effect on the propensity for developing a certain disease or characteristic. In contrast, complex traits, such as eye color or cardiovascular disease, are determined by variations occurring in many genes that have smaller effect sizes and act over long periods of time, often in concert with environmental factors. The cumulative risk derived from aggregating contributions of the many DNA variants associated with a complex trait or disease is referred to as a polygenic risk score (also known as a genetic risk score). This JAMA Genomics and Precision Health article explains polygenic risk scores as determinants of an individual’s inherited risk for complex disease.
Article
Importance Anxiety and stress-related disorders are among the most common mental disorders. Although family and twin studies indicate that both genetic and environmental factors play an important role underlying their etiology, the genetic underpinnings of anxiety and stress-related disorders are poorly understood. Objectives To estimate the single-nucleotide polymorphism–based heritability of anxiety and stress-related disorders; to identify novel genetic risk variants, genes, or biological pathways; to test for pleiotropic associations with other psychiatric traits; and to evaluate the association of psychiatric comorbidities with genetic findings. Design, Setting, Participants This genome-wide association study included individuals with various anxiety and stress-related diagnoses and controls derived from the population-based Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH) study. Lifetime diagnoses of anxiety and stress-related disorders were obtained through the national Danish registers. Genes of interest were further evaluated in mice exposed to chronic social defeat. The study was conducted between June 2016 and November 2018. Main Outcomes and Measures Diagnoses of a relatively broad diagnostic spectrum of anxiety and stress-related disorders. Results The study sample included 12 655 individuals with various anxiety and stress-related diagnoses and 19 225 controls. Overall, 17 740 study participants (55.6%) were women. A total of 7308 participants (22.9%) were born between 1981-1985, 8840 (27.7%) between 1986-1990, 8157 (25.6%) between 1991-1995, 5918 (18.6%) between 1996-2000, and 1657 (5.2%) between 2001-2005. Standard association analysis revealed variants in PDE4B to be associated with anxiety and stress-related disorder (rs7528604; P = 5.39 × 10⁻¹¹; odds ratio = 0.89; 95% CI, 0.86-0.92). A framework of sensitivity analyses adjusting for mental comorbidity supported this result showing consistent association of PDE4B variants with anxiety and stress-related disorder across analytical scenarios. In mouse models, alterations in Pde4b expression were observed in those mice displaying anxiety-like behavior after exposure to chronic stress in the prefrontal cortex (P = .002; t = −3.33) and the hippocampus (P = .001; t = −3.72). We also found a single-nucleotide polymorphism heritability of 28% (standard error = 0.027) and that the genetic signature of anxiety and stress-related overlapped with psychiatric traits, educational outcomes, obesity-related phenotypes, smoking, and reproductive success. Conclusions and Relevance This study highlights anxiety and stress-related disorders as complex heritable phenotypes with intriguing genetic correlations not only with psychiatric traits, but also with educational outcomes and multiple obesity-related phenotypes. Furthermore, we highlight the candidate gene PDE4B as a robust risk locus pointing to the potential of PDE4B inhibitors in treatment of these disorders.
Article
Background: Habitual alcohol use can be an indicator of alcohol dependence, which is associated with a wide range of serious health problems. Methods: We completed a genome-wide association study in 126,936 European American and 17,029 African American subjects in the Veterans Affairs Million Veteran Program for a quantitative phenotype based on maximum habitual alcohol consumption. Results: ADH1B, on chromosome 4, was the lead locus for both populations: for the European American sample, rs1229984 (p = 4.9 × 10-47); for African American, rs2066702 (p = 2.3 × 10-12). In the European American sample, we identified three additional genome-wide-significant maximum habitual alcohol consumption loci: on chromosome 17, rs77804065 (p = 1.5 × 10-12), at CRHR1 (corticotropin-releasing hormone receptor 1); the protein product of this gene is involved in stress and immune responses; and on chromosomes 8 and 10. European American and African American samples were then meta-analyzed; the associated region at CRHR1 increased in significance to 1.02 × 10-13, and we identified two additional genome-wide significant loci, FGF14 (p = 9.86 × 10-9) (chromosome 13) and a locus on chromosome 11. Besides ADH1B, none of the five loci have prior genome-wide significant support. Post-genome-wide association study analysis identified genetic correlation to other alcohol-related traits, smoking-related traits, and many others. Replications were observed in UK Biobank data. Genetic correlation between maximum habitual alcohol consumption and alcohol dependence was 0.87 (p = 4.78 × 10-9). Enrichment for cell types included dopaminergic and gamma-aminobutyric acidergic neurons in midbrain, and pancreatic delta cells. Conclusions: The present study supports five novel alcohol-use risk loci, with particularly strong statistical support for CRHR1. Additionally, we provide novel insight regarding the biology of harmful alcohol use.