This is the html version of the file https://kclpure.kcl.ac.uk/portal/files/226039250/2023_Ter_Kuile_Abigail_1439209_ethesis.pdf.
Google automatically generates html versions of documents as we crawl the web.
These search terms have been highlighted: cognitive behavioral therapy sexual women ter kuile 2010
Page 1
This electronic thesis or dissertation has been
downloaded from the King’s Research Portal at
https://kclpure.kcl.ac.uk/portal/
Take down policy
If you believe that this document breaches copyright please contact librarypure@kcl.ac.uk providing
details, and we will remove access to the work immediately and investigate your claim.
END USER LICENCE AGREEMENT
Unless another licence is stated on the immediately following page this work is licensed
under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International
licence. https://creativecommons.org/licenses/by-nc-nd/4.0/
You are free to copy, distribute and transmit the work
Under the following conditions:
Attribution: You must attribute the work in the manner specified by the author (but not in any
way that suggests that they endorse you or your use of the work).
Non Commercial: You may not use this work for commercial purposes.
No Derivative Works - You may not alter, transform, or build upon this work.
Any of these conditions can be waived if you receive permission from the author. Your fair dealings and
other rights are in no way affected by the above.
The copyright of this thesis rests with the author and no quotation from it or information derived from it
may be published without proper acknowledgement.
Leveraging genome-wide data to understand the risk and treatment of common mental
disorders
ter Kuile, Abigail
Awarding institution:
King's College London
Download date: 11. Nov. 2023

Page 2
Leveraging genome-wide data to
understand the risk and treatment
of common mental disorders
Abigail Roos ter Kuile
Social, Genetic and Developmental Psychiatry Centre
Institute of Psychiatry, Psychology and Neuroscience
King's College London
This thesis is submitted for the degree of
Doctor of Philosophy in Statistical Genetics
Supervised by Professor Thalia Eley and Professor Gerome
Breen
March 2023

Page 3
2
Abstract
Common mental disorders, including anxiety and depression, have a detrimental impact on
global disease burden and quality of life. Both the risk and treatment of anxiety and depression
are associated with the complex interplay between genetic, psychological, and socio-
environmental factors. Integrating these factors has posed a challenge in research efforts
aimed at improving the prevention and treatment of these disorders. The recent growth of
genomic data and advancements in multi-trait methodology allows for the incorporation of
genetics with psycho-social factors at an unprecedented level. This thesis applies statistical
genetic approaches to further understand the genetic influences on the risk and resolution of
common mental disorders. The first empirical study (Chapter 2) presents the largest genome-
wide association study (GWAS) meta-analysis of lifetime fear-based anxiety disorders (N total
=188,812; N cases =30,861) and examines the shared and distinct genetic relationship with
generalised anxiety disorder (N total =172,248; N cases =54,928) and broad domains of other
complex traits. Chapter 3 builds on previous research finding a genetic component of reported
trauma, a major socio-environmental risk factor for anxiety and depression. By leveraging pre-
existing GWAS summary statistics, genetic correlations and genomic multiple regression
analyses are used to identify heritable psychological and behavioural traits that capture the
common genetic variant-based heritability of reported childhood maltreatment (N=185,414).
Chapter 4 represents the largest GWAS meta-analysis of outcomes following psychological
treatment for anxiety and depression (N total =15,131; N reported positive outcomes =11,408).
The utility of genetic factors is also assessed by incorporating polygenic scores of complex
traits into multivariable prediction models of psychological treatment outcomes alongside
known clinical and demographic predictors. The final chapter discusses the implications of the
findings from this thesis within the context of challenges emerging in anxiety and depression
genomics. With the ongoing expansion of GWAS data, consideration should be taken into how
phenotyping approaches influence downstream analyses, including the genetic structure
observed across psychopathologies and the psycho-social components involved in gene-
environment interplay.

Page 4
3
Table of contents
Abstract ...................................................................................................................... 2
Table of contents ...................................................................................................... 3
List of tables and figures .......................................................................................... 5
Acknowledgments .................................................................................................... 7
Statement of authorship ........................................................................................... 8
Publications related to this thesis ......................................................................... 10
Chapter 1. General introduction ............................................................................ 12
Anxiety and depression comorbidity is extensive and associated with adverse outcomes
........................................................................................................................................... 12
Research into understanding drivers of comorbidity whilst accounting for clinical
heterogeneity ................................................................................................................. 14
Transdiagnostic and disorder-specific genetic influences ................................................. 16
Twin study findings ........................................................................................................ 16
From twin studies to genome-wide methods to understand biological aetiology ........... 19
Progress in depression and anxiety disorder genomics ................................................ 26
Leveraging genomic data to understand pervasive comorbidity and clinical heterogeneity
....................................................................................................................................... 28
Social environmental influences on anxiety and depression ............................................. 31
The heritability of environmental measures ................................................................... 32
Heritable psychological influences on anxiety and depression: from risk to treatment ..... 34
Heritable influences on subjective self-reports of adverse experiences ........................ 34
Heritable influences on positive experiences and application to psychological treatment
....................................................................................................................................... 35
Summary and aims ............................................................................................................ 38
References ........................................................................................................................ 39
Chapter 2. Exploring the genome-wide genetic overlap between anxiety and
fear disorders .......................................................................................................... 51
Abstract ............................................................................................................................. 52
Introduction ........................................................................................................................ 53
Methods ............................................................................................................................. 56
Results ............................................................................................................................... 62
Discussion ......................................................................................................................... 71
References ........................................................................................................................ 76
Chapter 3. Genetic decomposition of the heritable component of reported
childhood maltreatment ......................................................................................... 81
Abstract ............................................................................................................................. 82
Introduction ........................................................................................................................ 83
Methods and Materials ...................................................................................................... 86
Results ............................................................................................................................... 88
Discussion ......................................................................................................................... 93

Page 5
4
References ........................................................................................................................ 99
Chapter 4. Genetic influences on self-reported outcomes following
psychological treatment for depression and anxiety disorders ....................... 103
Abstract ........................................................................................................................... 104
Introduction ...................................................................................................................... 105
Methods ........................................................................................................................... 109
Results ............................................................................................................................. 117
Discussion ....................................................................................................................... 123
References ...................................................................................................................... 129
Chapter 5. General discussion ............................................................................ 134
General overview of findings ........................................................................................... 134
Theme 1: Phenotyping depth and the utility of self-reported measures .......................... 137
Theme 2: the genetic structure of internalising disorders and implications for disorder
grouping ........................................................................................................................... 139
Theme 3: interpreting GWAS findings in the context of psychosocial influences ............ 143
Personality, behaviour and cognitive biases ............................................................... 144
Socioeconomic status .................................................................................................. 146
General Limitations .......................................................................................................... 147
Lack of replication and sources of bias ....................................................................... 147
Lack of ancestral diversity ........................................................................................... 149
Future directions .............................................................................................................. 150
Establishing causality, and disentangling individual from familial genomic effects ..... 150
Expanding and integrating genome-wide data ............................................................ 152
Conclusions ..................................................................................................................... 153
References ...................................................................................................................... 154
Appendix A. Supplementary materials for Chapter 2 ........................................ 160
Appendix B. Supplementary materials for Chapter 3 ........................................ 211
Appendix C. Supplementary materials for Chapter 4 ........................................ 241

Page 6
5
List of tables and figures
Chapter 1
Figure 1: An overview of the three empirical chapters presented in this thesis and the
overlap among genomic, socio-environmental and psychological factors. ............................ 16
Box 1: Definitions and approaches to estimate trait heritability ............................................. 18
Box 2: Definitions of types and consequences of human genetic variation ........................... 20
Box 3: Structural properties of the human genome and related population genetic concepts
leveraged in genome-wide analyses ..................................................................................... 21
Figure 2: An illustration of the various stages of genome-wide analyses. ............................. 23
Box 4: Definitions of pleiotropy, bivariate and multi-trait genetic analyses ............................ 24
Figure 3: An illustration of the genomic continuum of the mood disorder spectrum from
mania genomics to internalising genomics. ........................................................................... 30
Figure 4: An illustration of the potential mechanisms underpinning the course and resolution
of common mental disorders, which are influenced by the interplay between genetic, socio-
environmental and psychological factors. .............................................................................. 31
Chapter 2
Table 1. Sample sizes in each study, GWAS dataset, and meta-analysis of fear-based
disorders and GAD. ............................................................................................................... 59
Figure 1: Genome-wide association study Manhattan plots for anxiety disorder phenotypes
meta-analysed across the GLAD+, QIMR and UKB datasets. .............................................. 63
Table 2: Independent genome-wide significant loci associated with anxiety disorder
phenotypes ............................................................................................................................ 64
Table 3: Gene-level associations with anxiety disorder phenotypes. .................................... 67
Table 4: Genetic correlations (rg ) between fear-based disorder and GAD phenotypes ....... 69
Figure 2: Genetic correlations between anxiety disorder phenotypes and external traits
estimated in LDSC regression. .............................................................................................. 70
Chapter 3
Figure 1. Top bivariate genetic correlations (rg) between reported childhood maltreatment
and various heritable traits. ................................................................................................... 89
Figure 2. Path diagrams representing results from genomic multiple regression analyses. . 91

Page 7
6
Chapter 4
Table 1. Number of participants from the GLAD+ and AGDS datasets with data on each
measures of self-reported outcomes following therapy (Total N = 15,131) ......................... 112
Table 2. Descriptive table for GLAD+ and AGDS datasets, total N = 15,131 ...................... 117
Table 3. LDSC SNP-based heritability estimates (using summary-level data) in GLAD+ and
AGDS datasets, total N = 15,131 ........................................................................................ 118
Table 4. GCTA-GREML SNP-based heritability estimates (using individual-level data) in
GLAD+ and AGDS datasets ................................................................................................ 118
Figure 1: Q-Q plot (left) and Manhattan plot (right) of associations from a meta-analysis of
retrospectively self-reported treatment outcomes following psychological therapy (N =
15,131; 75% reported positive outcomes). .......................................................................... 119
Table 5. Univariable associations between predictors and retrospectively self-reported
treatment outcomes in the GLAD+ dataset (N = 4,439) ...................................................... 121
Table 6. Models predicting self-reported outcomes following psychological therapy for
depression/anxiety using genetic, sociodemographic and clinical predictors in the GLAD+
dataset (N = 4,439) .............................................................................................................. 122
Chapter 5
Figure 1: An illustration of the emergence of three key themes based on the findings of this
thesis and the way they relate to various stages of GWAS. ................................................ 137
Figure 2: An illustration of the genetic continuum across the mood disorder spectrum to the
internalising disorder spectrum, with the addition of fear and GAD. .................................... 141

Page 8
7
Acknowledgments
Thank you to the study participants who shared their life experiences, and all the individuals
involved in designing and managing the studies and data, without whom the research
presented in this thesis would not have been possible. Additionally, I am thankful to the NIHR
Maudsley Biomedical Research Centre for providing me with the opportunity and funding to
pursue this PhD.
I am deeply grateful to my supervisors, Thalia and Gerome, for their unwavering support and
guidance. Your mentorship has provided me with numerous exciting opportunities to grow as
a researcher and has been instrumental in shaping my research and enabling me to achieve
my academic goals. To Thalia, thank you for your kindness, encouragement, and especially
your support during the challenging period when I was unwell with long covid. You gave me
invaluable insight to see the bigger picture of my research and communicate my findings more
broadly, which I will take with me throughout my career. To Gerome, thank you for helping me
find my passion and introducing me to the world of psychiatric genetics. You consistently had
faith in my ability to tackle challenges and encouraged me to persevere. I am thankful for the
opportunity to work with you both.
I feel fortunate to have had the opportunity to work at the SGDP Centre and meet so many
wonderful people. Despite the pandemic, the support and sense of community that I
experienced (even online) was truly special. I am grateful to my colleagues in the EDIT and
TNG research teams for being a part of my journey. Working alongside such a friendly,
sociable, and supportive group has been an enriching and rewarding experience.
A special thank you to Jess, Katie, Helena, Alicia, and Anna, whose friendship and
collaboration have been a highlight of my PhD journey. You all have been an inspiration to me
and have lifted me up during the most challenging times. Your support and encouragement
have helped me grow as a scientist and as a person, and I have learned so much from each
of you. Thank you for being there to celebrate every achievement and for encouraging me to
be as proud of myself as I am of each of you.
To my dear friends, and especially, my housemates at Greig and Sandlings, thank you for
taking care of me, reminding me to take breaks, and ensuring that I went outside to get some
fresh air during periods of intense focus. Your kindness and positivity were crucial in helping
me maintain my wellbeing.
I am also deeply grateful to my wonderful family - my Mum, Dad, and Naomi. Your unwavering
love, support, and inspiration have brought me to where I am today. We spent a considerable
portion of my PhD in lockdown together which I will always look back on fondly. Your care and
encouragement when I was unwell with long covid helped me to pace myself until I was back
to my normal self, enabling me to complete this hurdle.
Finally, to Oscar. I can’t thank you enough for everything. You have been my rock and
sounding board, providing me with love, encouragement, patience, and support every step of
the way. You have kept me grounded and reminded me that there is life outside of the PhD
when I needed it most. Thank you for being by my side throughout this journey.

Page 9
8
Statement of authorship
All the work presented in this thesis is my own except where acknowledged in the text. Data
collection for all study samples was completed by the respective research teams. Genetic data
quality control (QC) was carried out by respective studies. In Chapters 2 and 4, I conducted
additional QC in subsamples of the GLAD+ datasets (Genetic Link to Anxiety and Depression
(GLAD) study and the COVID-19 Psychiatric and Neurological Genetics (COPING) study).
During the duration of this PhD, I was involved in setting up and maintaining three pipelines in
collaboration with colleagues to generate repositories of data used in this thesis and for future
studies. First, I contributed to the phenotypic data cleaning pipeline of the GLAD+ dataset
(used in Chapters 2 and 4). Second, I helped maintain and update a GWAS summary
statistics QC pipeline and repository of hundreds of complex traits (used in Chapters 2, 3 and
4). Third, using the GWAS summary statistics from this repository, I contributed to setting up
a polygenic score repository in the GLAD+ dataset (Chapter 4).
The studies presented in Chapters 2 and 3 were conceived and conducted by me as first
author (A.R.T.K), in collaboration with colleagues included in the author list presented at the
start of each chapter. The work presented in Chapter 4 was a joint first author study conducted
with Alicia J. Peel, reflecting equal contributions from myself and A.J.P. I contributed to the
work in Chapter 4 by leading on genome-wide association analyses in the GLAD+ dataset,
GWAS meta-analysis across datasets, post-GWAS analyses, genetic quality control
procedures and additionally the creation of polygenic risk scores for prediction modelling, with
input from A.J.P. I assisted A.J.P who led on preparation of phenotypic data, prediction
modelling, preparing the pre-registration and an original draft of the manuscript. Chapters 2
and 4 include GWAS meta-analyses that were conducted in collaboration with researchers
involved in the Australian Genetics of Depression study (AGDS) based at Queensland Institute
of Medical Research Berghofer (QIMR). Analyses in the AGDS dataset were conducted by
researchers at the QIMR. I led on coordinating and generating scripts to harmonise data
analysis with AGDS collaborators, including phenotypic definitions and genome-wide analysis
scripts. A summary of chapter-specific author contributions is shown below.
Abigail ter Kuile

Page 10
9
Chapter 2
A.R.T.K, T.C.E and G.B. were responsible for the study conception and design. T.C.E, G.B,
G.K., M.R.D, M.H., N.R.W, S.E.M, E.M.B, N.G.M were responsible for the acquisition of the
data. A.R.T.K, C.H., A.J.P, H.L.D, J.M., Y.L., J.Z, A.P. were responsible for phenotypic data
preparation and cleaning. Preparation and quality control of genetic data was conducted by
A.R.T.K, S.H.L, J.R.I.C and B.A. B.M carried out data analysis in the AGDS QIMR dataset.
A.R.T.K conducted all other data analyses with support from G.M.V, S.H.L, H.L.D, J.M, M.S,
J.R.I.C and A.E.F. A.R.T.K was responsible for the drafting and revising of the manuscript,
under the close supervision of T.C.E. All co-authors reviewed the manuscript.
Chapter 3
A.R.T.K and G.B were responsible for the study conception and design, with input from all co-
authors. A.R.T.K, G.B, C.H, J.R.I.C, D.F.L, J.G and M.B.S were responsible for the acquisition
of the data. A.R.T.K conducted the analyses, with support from C.H. and C.R. A.R.T.K was
responsible for the drafting and revising of the manuscript, under the close supervision of G.B
and T.C.E. All co-authors contributed to the interpretation of the data, and reviewed and edited
the manuscript.
Chapter 4
A.R.T.K, A.J.P, C.R and T.C.E conceived and designed the study. T.C.E, G.B, G.K., M.R.D,
M.H., N.R.W, S.E.M, E.M.B, N.G.M were responsible for the acquisition of the data.
Phenotypic data in the GLAD+ dataset was processed and cleaned by A.J.P, A.R.T.K, C.H.,
H.L.D, J.M. Preparation and quality control of genetic data was conducted by A.R.T.K, S.H.L,
J.R.I.C and B.A. A.R.T.K led on genome-wide analyses, with support from J.M, M.S and
S.H.L. A.R.T.K generated polygenic scores in the GLAD+ dataset, with support from H.L.D
and Oliver Pain. A.J.P ran prediction modelling analysis, with support from A.R.T.K and Oliver
Pain. B.M and K.H carried out data analysis in the AGDS dataset. All authors contributed to
the interpretation of the results. A.J.P led on preparing an original draft of the manuscript
submitted in A.J.P’s PhD thesis, with input from A.R.T.K and supervisory input from T.C.E.
A.R.T.K was responsible for preparing and revising the current version of the manuscript
included in this thesis, with supervisory input from T.C.E and revised by G.B.

Page 11
10
Publications related to this thesis
Publications resulting from the chapters in this thesis
Chapter 3 has been accepted for publication following peer-review in the journal Biological
Psychiatry Global Open Science.
ter Kuile, A. R., H�bel, C., Cheesman, R., Coleman, J. R. I., Peel, A. J., Levey, D. F., Stein,
M. B., Gelernter, J., Rayner, C., Eley, T. C., & Breen, G. (In press). Genetic
decomposition of the heritable component of reported childhood maltreatment.
Biological Psychiatry Global Open Science.
https://doi.org/10.1016/j.bpsgos.2023.03.003
Chapter 2 and 4 are currently in preparation for publication.
Chapter 2:
ter Kuile, A. R., Mitchell, B. L., Morneau-Vaillancourt, G., Lee, S. H., Davies, H. L., Mundy,
J., Peel, A. J., Skelton, M., H�bel, C., Davies, M. R., Coleman, J. R. I., F�rtjes, A. E.,
Ahmad, Z., Lin, Y., Adey, B. N., McGregor, T., Purves, K., Palmos, A., Zvrskovec, J.,
Hotopf, M., Kalsi, G., Jones, I. R., Smith, D. J., Veale, D., Walters, J. T. R., Armour,
C., Hirsch, C. R., McIntosh, A. M., Wray, N. R., Medland, S. E., Byrne, E. M., Martin,
N. G., Breen, G., & Eley, T. C. (In prep). Exploring the genome-wide genetic overlap
between anxiety and fear disorders.
Chapter 3:
Peel, A. J.*, ter Kuile, A. R.*, Rayner, C., Hopkins, K., Mitchell, B. L., Medland, S. E., Lee, S.
H., Adey, B. N., Armour, C., Buckman, J. E. J., Byrne, E. M., Coleman, J. R. I., Danese,
A., Davies, M. R., Davies, H. L., Hickie, I. B., Hirsch, C., Hotopf, M., H�bel, C., Jones,
I. R., Kalsi, G., Krebs, G., Martin, N. G., McIntosh, A. M., Mundy, J., Purves, K. L.,
Rogers, H., Skelton, M., Smith, D. J., Veale, D., Walters, J. T. R., Wray, N. R., Breen,
G., & Eley, T. C.(In prep). Genome-wide analysis and prediction of self-reported
outcomes following psychological treatment for depression and anxiety.
* Joint first authors

Page 12
11
Additional publications
Co-authored publications in peer-reviewed journals arising from additional
work conducted during this PhD
Rayner, C., Coleman, J., Skelton, M., Armour, C., Bradley, J. R., Buckman, J. E. J., Davies,
M., Hirsch, C., Hotopf, M., H�bel, C., Jones, I. R., Kingston, N., Krebs, G., Lin, Y.,
Monssen, D., McIntosh, A., Mundy, J., Peel, A., Rimes, K., Rogers, H., Smith, D. J.,
ter Kuile, A. R, … Breen, G. & Eley, T.C., (2022). Patient characteristics associated
with retrospectively self-reported treatment outcomes following psychological therapy
for anxiety or depressive disorders - a cohort of GLAD Study participants. BMC
Psychiatry.
Young, K. S., Purves, K. L., H�bel, C., Davies, M. R., Thompson, K. N., Bristow, S., Krebs,
G., Danese, A., Hirsch, C., Parsons, C. E., Vassos, E., Adey, B. N., Bright, S.,
Hegemann, L., Lee, Y. T., Kalsi, G., Monssen, D., Mundy, J., Peel, A. J., Rayner, C.,
Rogers, H. C., ter Kuile, A. R., … , Eley, T. C. & Breen, G., (2022). Depression, anxiety
and PTSD symptoms before and during the COVID-19 pandemic in the UK.
Psychological Medicine.
Peel, A. J., Purves, K. L., Baldwin, J. R., Breen, G., Coleman, J. R. I., Pingault, J. B., Skelton,
M., ter Kuile, A. R., Danese, A. & Eley, T. C., (2022). Genetic and early environmental
predictors of adulthood self-reports of trauma. British Journal of Psychiatry.
Davies, M. R., Buckman, J. E. J., Adey, B. N., Armour, C., Bradley, J. R., Curzons, S. C. B.,
Davies, H. L., Davis, K. A. S., Goldsmith, K. A., Hirsch, C. R., Hotopf, M., H�bel, C.,
Jones, I. R., Kalsi, G., Krebs, G., Lin, Y., Marsh, I., McAtarsney-Kovacs, M., McIntosh,
A. M., Mundy, J., Monssen, D., Peel, A. J., Rogers, H. C., Skelton, M., Smith, D. J., ter
Kuile, A. R., … , Breen, G. & Eley, T. C., (2022). Comparison of symptom-based
versus self-reported diagnostic measures of anxiety and depression disorders in the
GLAD and COPING cohorts. Journal of Anxiety Disorders.
Peel, A. J., Armour, C., Buckman, J. E. J., Coleman, J. R. I., Curzons, S. C. B., Davies, M. R.,
H�bel, C., Jones, I., Kalsi, G., McAtarsney-Kovacs, M., McIntosh, A. M., Monssen, D.,
Mundy, J., Rayner, C., Rogers, H. C., Skelton, M., ter Kuile, A. R., Thompson, K. N.,
Breen, G., Danese, A., Eley, T. C., (2021). Comparison of depression and anxiety
symptom networks in reporters and non-reporters of lifetime trauma in two samples of
differing severity. Journal of Affective Disorders Reports.
Davies, H. L., H�bel, C., Herle, M., Kakar, S., Mundy, J., Peel, A. J., ter Kuile, A. R., … Breen,
G., (2022). Risk and protective factors for new-onset binge eating, low weight, and
self-harm symptoms in> 35,000 individuals in the UK during the COVID-19 pandemic.
The International Journal of Eating Disorders.
Davies, M. R., Glen, K., Mundy, J., ter Kuile, A. R., Adey, B. N., Armour, C., Assary, E.,
Coleman, J. R. I., Goldsmith, K. A., Hirsch, C. R., Hotopf, M., H�bel, C., Jones, I. R.,
Kalsi, G., Krebs, G., McIntosh, A. M., Morneau-Vaillancourt, G., Peel, A. J., Purves, K.
L., … Eley, T. C. (2023). Factors associated with anxiety disorder comorbidity. Journal
of Affective Disorders.

Page 13
12
Chapter 1. General introduction
Anxiety and depression comorbidity is extensive and associated with
adverse outcomes
Common mental disorders account for a significant proportion of the global disease burden1.
They comprise multiple anxiety and depressive disorders and are a leading cause of disability,
with a global lifetime prevalence of approximately 16% and 11%, respectively2–4. Anxiety, fear
and sadness are typical emotions experienced in everyday life, but when experienced at
pathological levels can interfere with day-to-day functioning and become debilitating5. The
impairing features of anxiety and depression lead to a widespread disability affecting broad
aspects of quality of life, including problems maintaining relationships, poor educational
attainment, unemployment and financial difficulties5–8.
In addition to the detrimental impact on quality of life, there is a strong economic case for
improving the prevention and treatment of common mental disorders. In the UK, anxiety and
depression contribute 17.5% (�20.5 billion) and 22.5% (�26.6 billion), respectively, to a
minimum annual cost of �117.9 billion arising from mental health problems. This equates to
5% of the total gross domestic product. These costs include the effect of mental health
problems on disability, occupational productivity and health services. Moreover, current UK
mental health services are under extreme demand and struggle to meet the needs of all those
who seek a diagnosis or treatment9. When left untreated, anxiety disorders and depression
are often experienced for extensive periods, becoming either chronic or recurrent, contributing
to a large proportion of suicide1,10–12.
Anxiety disorders are characterised by excessive fear and anxiety, accompanied by impairing
features such as avoidance behaviours, physical anxiety reactions and panic attacks. In the
Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5), specific clusters
of symptoms differentiate one anxiety disorder from the other and include generalised anxiety
disorder (GAD), agoraphobia, social anxiety disorder, specific phobia and panic disorder13.
The breadth and focus of the threat that causes anxiety or fear differs across the disorders.
General anxiety is the anticipation and pathological preoccupation with a range of future
potential threats. Essential criteria for a GAD diagnosis include experiencing general anxiety
persistently for at least six months and difficulties controlling the worry. In contrast, fear is an
emotional and physiological reaction to immediate threats. The particular focus of perceived
threat distinguishes the phobias from each other and must consistently be feared or avoided
for a minimum period of six months for a diagnosis. The focus of fear is the broadest in

Page 14
13
agoraphobia; the fear that escape might not be possible or help might not be available if
something were to go wrong in a range of situational exposures. In social anxiety disorder
(also known as social phobia), a narrower focus is placed on situations involving social
scrutiny. In specific phobias, the focus is limited to a certain situation or object that elicits fear.
A panic disorder diagnosis requires experiencing recurrent, unexpected panic attacks for at
least one month and consistent worry and avoidance of situations associated with having
panic attacks5.
Major depressive disorder (MDD) is the most common depressive disorder and has
considerable diagnostic heterogeneity. For a diagnosis of MDD, a low mood or the reduced
capacity to experience pleasure must be present and persist for at least two weeks, along with
the presence of other symptoms5. Clinical subtype specifiers can be symptom-based (with,
e.g. anxious-distress, mixed hypomanic, atypical, psychotic, melancholic features), temporal
or etiological-based (e.g. seasonal patterning, postpartum onset) and recurrence-based
(single episode or recurrent MDD)5,14.
Anxiety and depressive disorders are highly comorbid with one another, as well as other
psychiatric disorders and physical health problems. Estimates of comorbidity with another
mental health disorder are as high as 89% for anxiety disorders15 and approximately 75% for
major depressive disorder16,17. This includes comorbidity with a range of emotional and
behavioural psychopathologies, namely alcohol and substance use disorders, post-traumatic
stress disorder, obsessive-compulsive disorder, personality disorders and attention deficit
hyperactivity disorder5,15,18,19. However, the most common co-occurrence is among individual
anxiety disorders and depression. Approximately half of those who meet the diagnostic criteria
for one anxiety disorder have a history of another anxiety disorder10. Similarly, over half of
those with depression have at least one anxiety disorder and vice versa16,20,21.
In addition to anxiety and depression comorbidities being extensive, they are also associated
with various adverse clinical outcomes. A broad range of comorbid physical health problems
is common, including cardiovascular disease, diabetes, cancer, and irritable bowel
syndrome5,22–24. The presence of anxiety and depression is also linked with poor prognostic
outcomes in both chronic somatic diseases24 and, to some extent, treatment response in other
psychiatric disorders25. Compared to individuals with one psychiatric disorder, anxiety-
depression comorbidity is associated with increased symptom severity, impairment,
chronicity, the likelihood of suicide attempts, and is harder to treat20,21,26,27.

Page 15
14
Research into understanding drivers of comorbidity whilst accounting for
clinical heterogeneity
Due to the magnitude and clinical severity of anxiety-depression comorbidity, examining
factors driving shared liability has been a focus in decades of research28. Comorbidity can be
modelled for clinical and research utility by examining covariance patterns within a structural
equation modelling (SEM) framework. Modelling patterns of comorbidity highlight the
distinctive and shared features between disorders and indicate overlapping or unique liability
factors29. Such research has implications for improving diagnostic systems and understanding
transdiagnostic or disorder-specific risk factors and treatment targets. Previous research
examining shared liability underpinning comorbidity patterns has challenged diagnostic
categories and continues to do so today28,30,31. These studies have aimed to address the
problem of pervasive comorbidity whilst taking into account clinical heterogeneity.
The extensiveness of anxiety-depression comorbidity is reflected in well-replicated disorder-
based and symptom-based factor analyses. These have revealed a broad shared liability to
both anxiety and depression along with other disorders characterised by emotional symptoms
(e.g. PTSD, OCD and eating disorders). Anxiety and depressive disorders are often
conceptualised as being core to this internalising disorder dimension, thought to be driven by
shared negative affect31. Disorders outside of this dimension are differentiated by other core
components of psychopathology. For example, disorders characterised primarily by
behavioural difficulties comprise an externalising disorder spectrum (e.g. substance use
disorders, borderline personality disorder). Disorders characterised by disturbances in thought
content with delusions, hallucinations or dissociative symptoms form the thought disorder
spectrum (i.e. schizophrenia spectrum disorders)32–34. The placement of some aspects of
disorders varies, for example, neurodevelopmental disorders (e.g. ASD, ADHD) and mania
(i.e. bipolar disorders)35,36. As discussed above, anxiety disorders and depression are also
comorbid with disorders in other spectra. This is captured by an overarching psychopathology
liability dimension, known as the ‘p-factor’, which accounts for shared features and high rates
of comorbidity observed across psychiatric disorders37.
Within these spectra lie subfactors that represent disorder-specific clinical heterogeneity.
These subfactors reflect differences in core features and comorbidity patterns among the
specific disorders. For example, GAD has higher rates of comorbidity with depression than
other anxiety disorders17. A well-replicated structure in the internalising spectrum is splitting
anxiety disorders and depression into two highly correlated subfactors: distress and fear.
Distress-based disorders are mainly characterised by broad, pervasive negative emotionality

Page 16
15
(i.e. GAD is grouped with major depression). In contrast, fear-based disorders have more
context-specific features of distress, with fearful arousal and behavioural avoidance of a
narrower range of stimuli (i.e. agoraphobia, social anxiety disorder, specific phobia and panic
disorder)31,38. The splitting of internalising into distress and fear subfactors suggests an
overarching shared liability and some subfactor-specific risk factors that are not shared across
distress-fear dimensions.
The complex aetiology of common mental disorders poses a challenge to understanding what
drives comorbid psychopathologies and within-disorder heterogeneity. Both the risk and
treatment of anxiety and depression are associated with a broad range of genetic,
psychological and socio-environmental factors11,13. Such factors on their own are not enough
to cause the disorders or explain why some individuals respond better to certain treatments
than others. Furthermore, they do not act independently of one another, with a complex, multi-
layered interplay across different types of biological, psychological and social/environmental
factors. The interplay between these influences is dynamic, varying across the lifespan, from
person to person and across different psychiatric disorders39. Thus, resolving heterogeneity
and comorbidity problems in psychiatric research requires not only data on a broad range of
psychopathologies, but also a multi-level approach that integrates genetic, psychological, and
environmental factors.
In the next sections of this thesis introduction, progress in research of each facet of the bio-
psycho-social approach in the risk and treatment of anxiety and depression will be described.
By leveraging the complex interplay between genetic, environmental and psychological
influences, a focus will be placed on how genomic data can be used to understand each facet
and how this thesis aims to address key gaps in anxiety and depression genomics research,
depicted in Figure 1. A glossary of key statistical genetic terms and concepts relevant to this
thesis is provided in Boxes 1-4.

Page 17
16
Figure 1: An overview of the three empirical chapters presented in this thesis and the
overlap among genomic, socio-environmental and psychological factors.
Through the application of genome-wide data, the overlap between genomic and
psychosocial factors can be harnessed to better understand risk factors and treatment
outcomes for common mental disorders.
Transdiagnostic and disorder-specific genetic influences
Twin study findings
Pivotal findings from family and twin studies formed the basis for understanding genetic
influences on anxiety and depression, which paved the way for future genomic studies.
Research dating to the early-mid 20th century showed that anxiety and depressive disorders
aggregate in families. A four to six-fold increased risk of anxiety disorders and a three-fold risk
of depression were observed in individuals with an affected first-degree relative compared to
unaffected controls40,41. Co-aggregation across individual anxiety disorders and depression
was also observed42.
Twin studies confirmed that the familial aggregation of anxiety and depressive disorders is
largely due to heritable factors and, to a lesser extent, environmental factors40–42. Definitions
Chapter 2: What
genomic factors
contribute to
subtype
heterogeneity?
Chapter 3: What
genomic factors
contribute to
environmental
vulnerability?
Chapter 4: What
genomic factors
contribute to
psychological
treatment
outcomes?
Common
mental
disorders
Biological
e.g. genotype
Social
e.g. adverse
environments
Psychological
e.g. poor
response to
psychological
treatment

Page 18
17
and approaches to estimate heritability, including twin study estimates, are provided in Box 1.
Twin study heritability estimates of anxiety and depressive disorders are moderate, ranging
from 30-50%40–43. While substantial, these estimates are lower than rarer psychiatric disorders
such as schizophrenia and bipolar disorder (65-80%)43,44. Family and twin studies largely
support that familial risk and heritability estimates are similar across anxiety and MDD
subtypes; however, some find earlier-onset and recurrent MDD show greater familial risk and
heritability40,42,43,45. Twin studies have found that heritability estimates of anxiety and
depression measures are broadly similar across age groups43, although some report lower
heritability for childhood than adult depressive symptoms46. Stable genetic influences may
largely explain the moderate, long-term stability of anxiety and depressive symptoms across
the lifespan47,48. Furthermore, both stable and time-specific genetic factors are shared among
anxiety and depressive disorders49. These findings highlight how shared genetic influences
on anxiety and depression risk are important across the lifespan.
A major finding from twin studies is that high levels of shared genetic influences largely explain
the high comorbidity across anxiety and depressive disorders and related symptom
dimensions. Several twin studies found that GAD and MDD overlap almost entirely in genetic
variance50. A high genetic overlap across individual anxiety disorders is also consistently
greater than disorder-specific genetic influences42. Twin models of genetic covariation have
shown substantial shared genetic liability among anxiety and depression in childhood,
adolescence and adulthood50–53. A broad, internalising genetic factor is well-replicated across
twin datasets, supporting the overarching phenotypic structure of the internalising spectrum
discussed earlier51,53–55. The high genetic overlap between neuroticism, anxiety and
depression indicates genetic influences on shared negative affectivity drive internalising
psychopathology and anxiety-depression comorbidity56. Twin studies have further explored
the genetic relationship between anxiety and depression and disorders outside of the
internalising spectrum, showing pervasive genetic overlap with practically all forms of
psychopathology42,50. This overlap indicates cross-disorder genetics and the genetic factors
structure across disorders can be modelled as an overarching ‘p-factor’ liability that drives
comorbidity57,58. Studies also support partitioning psychiatric disorders into two distinct but
correlated lower-order internalising and externalising clusters59. As such, compared to their
overlap with other disorders, the genetic overlap is highest between anxiety and depression.
Notably, some multivariate twin modelling also supports the distress-fear subfactors structure
within the internalising dimension51,60,61. Multiple studies find that the extent of genetic overlap
between individual anxiety disorders and depression varies50. GAD and MDD show the
highest genetic overlap and are core to the internalising factor, indicating distress is a key

Page 19
18
driver of general emotional problems59. Panic disorder and phobias have robust genetic
correlations with distress but also show some genetic distinction from GAD and MDD, forming
a genetic liability fear factor51,60,61. Together, twin study findings indicate that grouping
disorders is appropriate to identify higher-order genetic influences that broadly influence
internalising. However, genetic studies that group distress with fear disorders may also miss
important sources of heterogeneity and insight into mechanistic differences.
Box 1: Definitions and approaches to estimate trait heritability
Heritability: The proportion of total phenotypic variance observed within a population (VP)
explained by genetic factors, with estimates ranging from zero to one. Heritability estimates
of a trait can vary across time and populations due to differences in, for example,
environmental contexts, the type of phenotypic instrument and age at measurement62,63.
Broad-sense heritability: Defined as H2 = VG / VP with genetic values (VG) attributed to both
additive and non-additive genetic factors. Non-additive influences include interactions
between alleles located within the same genetic locus (dominance) or located on different
loci (epistasis)62–64.
Narrow-sense heritability: Defined as h2 = VA /VP with genetic values (VA) attributed only to
additive genetic factors; the sum of the average effect of multiple alleles contributing to a
trait62–64.
Twin study heritability: Twin studies are a commonly used family-based approach to
estimate trait heritability and environmental components. Phenotypic differences between
genetically identical monozygotic twins (MZ) and ~50% genetically similar dizygotic twins
(DZ) are compared. Traits that are more phenotypically similar in MZ than in DZ twins indicate
the role of genetic variation. Variance components modelled in twin designs include additive
genetics (A), non-additive genetics (D), the individual environment not shared among sibling
pairs (E), and the common environment shared among siblings growing up in the same
family, thus accounting for some within family resemblance (C). In the classical ACE twin
model, narrow-sense heritability is estimated based on the assumption that the non-additive
effects are zero. In the ADE model, the C component is replaced to estimate D, enabling
broad-sense heritability to be calculated from the combined variance of A and D64,65. Twin
studies have found stronger evidence for the role of additive genetic influences on complex
traits than non-additive43,66.
SNP-based heritability (h2
SNP): The proportion of phenotypic variance explained by a given
set of genome-wide common genetic variants67.
GREML h2
SNP: Genomic relatedness restricted maximum likelihood (GREML) is a commonly
used method to estimate narrow-sense h2
SNP using individual-level genomic data in unrelated
individuals68. GREML is implemented in the software GCTA69. Genome-wide common SNPs
are leveraged to construct a genomic relatedness matrix (GRM) containing the relatedness
coefficient between each pair of individuals across each SNP. The extent to which genomic
similarity can predict pair-wise phenotypic similarity for a given trait is used to deconstruct
trait variance into additive genetic variance and residual variance components (e.g. non-
additive genetics, environment, error)64,67.

Page 20
19
Box 1: Definitions and approaches to estimate trait heritability
LDSC regression h2
SNP: Linkage disequilibrium score (LDSC) regression is a method to
estimate narrow-sense h2
SNP using GWAS summary statistics. LDSC regression assumes
under a polygenic model that common variants in higher LD regions are, on average, more
likely to be tagged by a causal variant in GWAS and therefore are more likely to be associated
with a given trait. An LD score quantifies the level of tagging of variants. A higher score
corresponds to a variant that tags more causal variants than those with a lower score and,
therefore, on average, has a higher association test statistic. LD scores are created using a
population reference panel representing the LD structure. For accurate estimates, the
population must be well-matched to the ancestry of the target GWAS population. An estimate
of h2
SNP is quantified as the regression coefficient from regressing the observed mean chi-
square association test statistic (Χ 2) against the LD score of each variant. The intercept can
be used to quantify bias from population stratification but can arise from an increase in sample
size and heritability of a trait. The attenuation ratio ([intercept -1]/ [Χ2 -1]) provides an estimate
of inflation not due to polygenicity, with a higher estimate indicating confounding67,70,71.
Liability scale h2
SNP: Observed scale h2
SNP estimates of categorical traits are converted to
the liability scale by accounting for the assumed population prevalence. The liability threshold
model presumes that genetic factors influence disorders under a normal distribution. An
individual meets case disorder status once reaching above a certain threshold of liability (on
the upper tail end of the normal distribution). The proportions of affected (cases) and
unaffected individuals (controls) in a GWAS sample often do not reflect the incidence of a
disorder observed in the population. The observed h2
SNP scale is often lower than the liability
h2
SNP scale, as information is lost by dichotomising the trait62,72.
From twin studies to genome-wide methods to understand biological aetiology
The heritable component and pervasive genetic overlap between anxiety and depressive
disorders showed the potential for employing genetic techniques to unravel biological
aetiology. Initial attempts to identify the genes associated with anxiety and depression
involved candidate gene and linkage analysis studies. As with all complex traits, these failed
to identify robust associations with anxiety and depression that survived replication42. As
knowledge of the human genome expanded, it was soon realised that psychiatric and other
complex traits are affected by many genetic variants, each of small effect size. In an attempt
to uncover this polygenicity, the era of genome-wide association studies (GWAS) began73.
A period of accelerated progress in genomics that led to the development of GWAS began
with the completion of the first human genome sequence, revealing millions of genetic
variants74,75. Box 2 provides a description of the various forms of human genetic variation.
Single nucleotide polymorphisms (SNP) are the most extensively analysed in GWAS, followed
by short insertion-deletion variants or ‘indels’. The majority of variants analysed in GWAS are
located within non-coding sequences of genes or in intergenic regions, which are potentially
involved in gene regulation76–78.

Page 21
20
Box 2: Definitions of types and consequences of human genetic variation
Allele: Alternative versions of base pair changes of a variant79.
Polygenic: A phenotype influenced by many genetic variants79. Each genetic variant
contributes to phenotypic variation with a small effect size. The sum of each small effect,
along with environmental influences, contributes to the continuous distribution of complex
trait variation observed in a population80.
Polymorphism: Multiple definitions of genetic polymorphism exist in the literature81. Here,
polymorphism is defined as a population-specific term related to the occurrence of two or
more alleles at a position in a DNA sequence, with the rarer allele occurring in at least 1% of
a given population82–84.
Single nucleotide variant: Variation from a reference genome at a single base pair change.
The most abundant form of genetic variation in the human genome76,77.
Single nucleotide polymorphism (SNP): A single nucleotide variant with the minor allele
present in at least 1% of the population77,85.
Insertions and deletions (indels): A variant that corresponds to the insertion or deletion of
one or more base pairs not found in the reference genome. The second most abundant form
of genetic variation in the human genome76,77.
Copy number variation: An intermediate-large scale form of structural variation,
corresponding to additional copies (duplications) or deletions of segments of sequences >
1,000 base pairs85,86.
Tandem repeats: A segment of sequences with a number of base pair repeats. Variants
with repeat sequences of 2-6 bases are termed short tandem repeats, and those 7-10 bases
are termed variable number tandem repeats77.
Synonymous and non-synonymous variants: SNVs, and to a lesser extent SNPs, can
occur in protein-coding sequences within genes (exons), and cause a change in the amino
acid sequence, termed a non-synonymous variant. In contrast, variants within exons that do
not change the amino acid sequence are termed synonymous variants77.
Missense variant: A type of non-synonymous variant that alters one amino acid in the
protein, potentially influencing protein function77.
Nonsense variant: A type of non-synonymous variant that results in a premature STOP
codon and an incomplete and potentially non-functional protein product77.
Intergenic variant: A variant located between genes77.
Intronic variant: A variant that falls within an intron - the non-coding sequences of genes77.
Since the completion of the first human genome sequence, technological advancements have
provided cost-effective tools to rapidly conduct GWAS77. As the number of sequenced human
genomes increased, the depth and detail of publicly available reference genomes
improved87,88. Such advancements have led to an improved understanding of the structural
properties of the human genome and population genetic concepts that are leveraged for
genome-wide analyses (Box 3). The cost of generating genotyping data for GWAS through
microarrays has decreased substantially over the years. It is the most commonly used
approach to gather genotype data as it costs approximately ~95% less than next-generation
sequencing89. Reference genomes are then used for imputation to expand the number of
variants detected from microarrays, improving the overlap of variants across datasets using

Page 22
21
different microarray technologies, thereby facilitating data harmonisation. The millions of
variants generated by these approaches are used in GWAS to conduct a hypothesis-free
study by testing across the entire genome if the frequency of alleles differs between cases
and controls (e.g. an allele is more common in individuals with anxiety disorder than in those
without), or along a trait dimension (e.g. an allele is associated with a higher GAD symptom
score)77. Variant associations may be causally or non-causally related to phenotype
expression, but are indirectly detected in GWAS due to being correlated with nearby causal
variants (in linkage disequilibrium [LD]). Haplotype blocks with regions of high LD are
leveraged to identify causal variants without the need for genotyping all causal SNPs90–92.
However, interpreting GWAS results is complicated by LD, giving rise to multiple associated
variants within a region, and subsequent analyses are required to determine causal
associations92–94.
Box 3: Structural properties of the human genome and related population genetic
concepts leveraged in genome-wide analyses
Reference panel: A database of densely genotyped haplotypes measured in a given set
of individuals from a specific population. Used to represent structural properties of the
genome in a given population, including allele frequencies and LD patterning. Examples of
commonly used reference panels include the Haplotype Reference Consortium, 1000
Genomes Project, and TOPMed95.
Allele frequencies: Allele frequency is the incidence of a certain allele in a population. In
a given population, minor allele frequency (MAF) is the frequency of the less common
allele, while major allele frequency is the frequency of the more common allele of a variant.
MAF is used to distinguish common from rare variants, with MAF > 1-5% considered
common79,90.
Linkage disequilibrium (LD): The non-random association between alleles located on
two distinct loci in a given population. Linkage equilibrium (LE) implies independence, in
which alleles at two loci are randomly associated by chance, with a theoretical
disequilibrium coefficient of 0. When the frequency of association between alleles at two
different loci deviates from expected when in LE, they are considered to be in LD. Levels
of LD are dependent on recombination events during meiosis that occur across
generations, natural selection, population bottlenecks, genetic drift, mutations, and
genomic inversion. LD can occur at a short-range, whereby LD is higher among physically
nearby genetic variants that are less likely to be separated by recombination events than
more distant variants. LD can occur at a long range, whereby the correlation between
genetic variants occurs at a larger distance, caused by various population processes91,96.
Haplotype block: Across the human genome are chromosomal regions with block-like
patterns of different LD levels. Low LD regions exist in recombination hot spots. In between
these hotspots are haplotype blocks, regions with high LD (from a few kb-100kb) and low
recombination, resulting in the inheritance of single-unit haplotype blocks across
generations. Haplotype blocks, therefore, reflect population history and geographical
subdivisions, with block sizes varying across ancestral populations91.
Genotype imputation: A statistical method that estimates genotypes of individuals at
unmeasured variants based on data from a more densely-genotyped population-specific
reference panel95,97.

Page 23
22
Box 3: Structural properties of the human genome and related population genetic
concepts leveraged in genome-wide analyses
Genetic ancestry group: A label to denote groups of individuals who share more similar
genetic ancestors. Genetic ancestry is estimated in GWAS participants by comparing
genotypes with global reference panels. A continuum of genetic differences across
ancestry populations arises as those with more common ancestors have more similar
genomic structural patterns due to a number of influences. This includes ancestors mating
within nearby geographic locations and the sharing of more recombination events than
those of more distant ancestors. These differences confer variable frequencies of alleles
and patterning of linkage disequilibrium across ancestry populations. It is, therefore,
common practice to limit GWAS to more homogenous genetic ancestry groups. This
reduces false-positive associations arising from allele-frequency differences across
populations instead of reflecting phenotypic variation. It should be noted that ancestry
groups are an oversimplification of the complex dimension of human genetic variation and
demography95,98.
Population structure and stratification: Population structure is the genetic structure (i.e.
LD and allele frequencies) of an underlying population due to non-random mating and
restricted geographical movement. When a phenotype is correlated with a population
structure in a sample, this gives rise to population stratification that can confound genome-
wide analyses95.
Principal component analysis (PCA): A method used to estimate population structure
within a GWAS dataset. PCA reduces the dimensions of large-scale data (i.e. millions of
variants measured across thousands of individuals) into components that capture a large
proportion of variance, reflecting the underlying structure of a dataset. PCs are used to
estimate and assign participants to genetic ancestry groups by comparing the data with a
population reference panel of known ancestries. Additionally, including PCs as covariates
in genome-wide analyses is a commonly used approach to control for population
stratification95,99,100.
Genetic variants tested in GWAS are typically restricted to alleles more common in a given
population, with a minor allele frequency of at least 1-5%76,90. This approach has shown that
many common variants of small effect size spread across the genome contribute to the genetic
liability of complex traits and are, therefore, highly polygenic101. Consequently, large sample
sizes are required to detect robust associations of small effect sizes across the millions of
tested variants. To account for this multiple testing burden, a Bonferroni correction for one
million independent statistical tests (5 x 10-8) is applied to establish a genome-wide
significance threshold and reduce the chance of false positives. An approximately linear
relationship exists between the number of genome-wide significant associations and sample
size73,102.
Large, well-powered GWAS are pivotal to understanding the aetiology of complex traits.
Summary results can be made publicly available and used for various downstream analyses
(Figure 2), including elucidating the biological underpinnings of complex traits through
systems biology approaches. GWAS summary results can be integrated with various
functional genomic data types, including features from cells and tissues, such as gene
expression data. The small effect of common genetic variant associations can be aggregated

Page 24
23
to the level of the gene and then combined into sets and translated to biological functions and
pathways enriched with a phenotype103.
Figure 2: An illustration of the various stages of genome-wide analyses.
Key population genetic concepts are applied to genome-wide association analyses (GWAS),
as described in Box 3. Summary results from GWAS can be used for several downstream
analyses. GWAS summary statistics are often made publicly available, enabling data on
various complex traits to be integrated into multiple-trait (e.g. multivariate or multivariable)
analyses. PC = principal component, LD = Linkage disequilibrium.
In addition to uncovering the genomic mechanisms associated with complex traits, GWAS
data can be used to estimate narrow-sense heritability captured by common genetic variants,
most often termed SNP-based heritability (see Box 1)67. Estimates of SNP-based heritability
can guide researchers on the sample size and power required to detect genome-wide
significant loci and the potential for future risk prediction based on GWAS data70. More
prevalent and highly polygenic psychiatric disorders with a lower SNP-based heritability, such
as anxiety and depression, require larger sample sizes than more heritable, rarer disorders,
such as schizophrenia, to achieve sufficient statistical power104. Estimates of SNP-based
heritability can be calculated directly from individual-level data used in GWAS, or through
summary-level GWAS results. As individual-level-based methods use full SNP data instead
of summary scores, they tend to yield higher estimates but are limited in computational speed,
and load with large sample sizes64,67. Using GWAS summary results is an alternative,
computationally efficient approach to estimating lower bounds of SNP-based heritability. This
has the additional benefit of not requiring access to individual-level genetic data for GWAS
datasets, often not possible in large-scale, collaborative meta-analyses. A widely used
summary-level method is linkage disequilibrium score (LDSC) regression (Box 1), which
Phenotyping
strategy
GWAS
SNP-based
heritability
genetic
correlations
polygenic
scoring
Multi-trait
analyses
Application of key concepts:
e.g. LD patterning, population structure
Population assignment
Genotype imputation
PC covariates

Page 25
24
exploits the fact that some SNPs are more correlated with each other than others (i.e. are in
higher LD, as described in Box 3)67,70,71.
A genetic correlation between two traits can be estimated using extensions of the same
approaches used to estimate SNP-based heritability. To estimate genetic correlations across
a wide range of complex traits, the computationally light and flexible approach of using GWAS
summary statistics in LDSC regression is common. LDSC regression estimates global (Box
4) correlations by estimating genetic covariation across a pair of phenotypes (as captured by
common genetic variants) and scaling by genetic variances (i.e. from SNP-based
heritability)105,106. A significant global genetic correlation between two phenotypes arises when
the direction of the effect of common genetic variants is consistent across the genome105.
When genetic effects in both traits are, on average, in the same direction, this results in a
positive correlation of up to 1, or when in the opposite direction, a negative correlation of up
to -1107. Of note, genetic correlations cannot be used to determine causality but can be used
to generate novel hypotheses about the relationship between two traits to be then tested in
follow-up analyses.
Box 4: Definitions of pleiotropy, bivariate and multi-trait genetic analyses
Pleiotropy: When a genetic variant (or locus, gene) is associated with more than one
trait106.
Horizontal pleiotropy: When a genetic variant influences two traits, either directly or
indirectly, through an additional intermediate phenotype. Indicative of shared biological
processes between two traits (also termed biological pleiotropy)106.
Vertical pleiotropy: When a genetic variant influences one trait, which in turn is causally
associated with a second trait (i.e. one trait mediates the effect of the genetic variant on
another trait). Reverse causation of a causal cascade between two phenotypes is also
possible. The presence of vertical pleiotropy can be useful for intervention purposes106.
Environmentally mediated pleiotropy: A type of vertical pleiotropy when a genetic variant
influences a phenotype that shapes the environment, which in turn influences a second
phenotype108.
Spurious pleiotropy: When a genetic variant is falsely associated with two traits. This can
arise due to multiple different sources of biases. For example, design artefacts such as
phenotype misclassification or ascertainment bias. High LD can also lead to spurious
pleiotropy by a variant tagging multiple causal variants located in different genes with
different functions, which are not causally related to both phenotypes106,109. Variants in
regions of extreme LD (e.g. the major histocompatibility complex) are sometimes removed
for genetic correlation analyses110.
Global genetic correlations: A bivariate global genetic correlation between two traits is
the average effect of pleiotropy across the whole genome106.
Local genetic correlations: A bivariate local genetic correlation between two traits
analyses region-specific pleiotropy (e.g. at a given locus). Different regions may show
genetic correlations in opposite directions, contributing to a non-significant global genetic
correlation111.

Page 26
25
Multi-trait analyses: Pervasive pleiotropy observed across multiple complex traits can be
leveraged through multi-trait analyses112–114. For example, multivariate genetic analyses (of
more than one outcome phenotype) can be used to extend bivariate genetic correlations
into multivariate Genomic Structural Equation Modelling115. Multivariable genetic analyses
(of one outcome phenotype and multiple predictors) can be used to model multiple
polygenic scores to predict a trait outcome jointly113,116.
A significant global genetic correlation can reflect multiple forms of underlying mechanisms
(Box 4). They may capture variants with a causal effect on multiple disorders, indicating
shared biological pathways70,106. Genetic correlations between psychiatric disorders and
health-related lifestyle factors may reflect causal relationships70. Genetic correlations
observed between personality and psychiatric disorders may be environmentally mediated108.
As such, measuring genetic correlations from GWAS across broad domains of phenotypes is
useful for understanding not only shared biological aetiology but also other sources of genetic
signals107.
To further strengthen hypotheses on shared and distinct mechanisms, a genetic overlap
across phenotypes can be harnessed for several downstream multi-trait analyses (Box 4).
Genetic correlations can be used to assess the genetic structure of traits related to psychiatric
disorders through multivariate modelling of genetic covariances. One example is the software
Genomic Structural Equation Modelling (Genomic SEM), which extends multivariate twin
modelling to genomic data using GWAS summary statistics115. This method constructs
estimates of SNP-based heritabilities and genetic correlations into matrices, often calculated
in LDSC regression. The genetic covariance matrix contains SNP-based heritabilities of each
trait and genetic correlations scaled relative to the heritabilities. A second sampling covariance
matrix contains the precision of these estimates (i.e. the sampling variance of each estimate
in the genetic covariance matrix), and the association between sampling errors to account for
GWAS sample overlap. A number of different structural equation models can then be fit to
these matrices. For example, this method can be used to evaluate hypotheses on why a group
of phenotypes are correlated or the extent to which other traits explain the genetic variance of
a trait in a model, or are unique to that phenotype115.
An alternative approach that leverages a genetic overlap between traits to build multi-trait
models is through polygenic scoring113. Polygenic scores are an aggregate of the number of
phenotype-associated alleles an individual carries. For example, for common mental
disorders, both individuals affected and unaffected by a disorder will carry a range of alleles
across the genome associated with an increased risk (coded as a 0, 1 or 2). To calculate a
polygenic score, alleles are summed and weighted by the effect size estimate of their
association with a given phenotype, derived from GWAS summary statistics in an independent

Page 27
26
sample117. A polygenic score for one trait can be used to explain the phenotypic variance of
another trait. This is particularly useful for examining the genetic overlap between two traits
where one trait does not have a well-powered GWAS available for genetic correlation analysis.
Even for polygenic scores derived from well-powered GWAS, the amount of phenotypic
variance explained is relatively small73,102. For example, one of the most powerful psychiatric
polygenic scores is for schizophrenia, which explains 8.5% of the variance in schizophrenia
liability118. As such, polygenic scores are not yet sufficiently powered for prediction at the
individual level. Prediction at the group level can be improved by combining multiple well-
powered polygenic scores and non-genetic predictors into multivariable models. As GWAS
sample sizes grow, the hope is that the power of polygenic scores will improve to the extent
that multivariable predictive models can be implemented for clinical application119.
Progress in depression and anxiety disorder genomics
Since the start of the GWAS era approximately 16 years ago, significant progress has been
made in psychiatric genomics research120. Collaborative efforts and large-scale data collection
have enabled sample sizes to reach the level of power needed to start identifying the
abundance of common genetic variants associated with psychiatric disorders. This has been
facilitated by the Psychiatric Genomics Consortium (PGC), where large-scale meta-analyses
of multiple GWAS have transformed psychiatric genomics research. Large national cohorts
such as the UK Biobank and the Million’s Veteran Programme (MVP) have also been crucial
in these developments by contributing data to the largest GWAS of depression and anxiety to
date45,121,122.
Considerable progress has been made in depression genomics and led to a broad array of
findings on the neurobiology of depression. The largest MDD GWAS identified 178
independent loci in a sample of approximately 370,000 cases123. The translation of depression
GWAS results have highlighted the role of several brain regions, nervous system
development, synaptic processes, and immune-related function123–126. Such findings have
progressed our understanding of the aetiology of depression by corroborating previous
hypotheses and generating new ones.
Anxiety disorder genomics, and thus an understanding of genomic-associated systems
biology, is somewhat behind compared to depression. A lack of datasets with measures on
the multiple types of anxiety disorders has hindered progress. This may partly be due to the
complexity of phenotyping anxiety, a highly comorbid trait with somewhat vague boundaries
between ‘normal’ to pathological states127,128. The largest GWAS of anxiety identified five
genome-wide significant loci associated with GAD symptoms in 175,000 individuals129. Two

Page 28
27
additional loci were associated with a case-control phenotype of anxiety and panic disorder in
34,000 cases129. A follow-up study applied a novel phenotyping approach to increase
statistical power by imputing GAD symptoms, identifying a further seven independent loci.
Integrating these GWAS results with functional genomic data found associations with the
dorsolateral prefrontal cortex and GABAergic neurons130. Although these studies represent
significant progress, larger datasets are still needed to further our understanding of the
genomic mechanisms underpinning anxiety disorders. Efforts are underway to publish the first
large-scale PGC anxiety disorder GWAS, representing a milestone in anxiety disorder
genomics127.
The SNP-based heritability estimates for anxiety and depression are broadly similar but are
also notably smaller than twin study estimates. Individual-level methods report ~26% SNP-
based heritability for any anxiety disorder131, and ~20% for MDD132,133. The ‘missing’ heritability
between SNP-based heritability and twin heritability estimates of complex traits are thought to
be explained by the role of genetic factors contributing to heritability not measured in GWAS
data, such as rare variants, non-additive genetics or gene-environment interactions73. As
noted earlier, summary-level SNP-based heritability estimates are even smaller than those
calculated from individual-level SNP data and twin data. The LDSC SNP-based heritability
estimate for MDD is 10.5%70,124 and 9% for broad depression125. Similarly, the largest GWAS
of anxiety reported an LDSC SNP-based heritability of 8.8%134, which is in line with other
anxiety disorder GWAS 135 and neuroticism GWAS136. However, meta-analysis SNP-based
heritability estimates of psychiatric disorders may also be biased downwards due to incorrect
conversions to the liability scale, not taking into account cohort-specific ascertainment (e.g.
an increase from 10.5% to 11.5% was observed for MDD)137. Furthermore, smaller, single-
study GWAS of anxiety disorders reported LDSC SNP-based heritability estimates as high as
28%138. This highlights how SNP-based heritability can vary across populations and
phenotypic measures73. Such heterogeneity across samples may, in part, limit the ability to
detect SNP-based heritability and contribute to missing heritability102.
As GWAS of anxiety and depression increase, a growing issue in the field is the balance
between increasing sample size and consistent and detailed phenotyping121,139,140. Many
large-scale genetic datasets use brief phenotypic measures, often limited to a few self-report
items to assess a broad psychiatric phenotype, as this approach is time and cost-effective.
Brief phenotyping has been key for making progress in loci discovery for both anxiety and
depression and will likely be vital for future risk prediction using GWAS data. The predictive
power of depression polygenic scores derived from GWAS is more dependent on increased
sample size than the detail of phenotyping. This indicates that sample size should be

Page 29
28
prioritised over phenotyping depth for risk estimation141. However, when GWAS sample sizes
are equal, polygenic scores for more detailed measures have captured more specific genetic
influences on MDD141,142. Loci identified by GWAS of briefly phenotyped broad depression are
also less specific to MDD140,142. A tradeoff thus exists between maximising sample size and
the depth of phenotyping.
While research on the depth of GWAS phenotyping for anxiety disorders is lacking, lessons
learnt from phenotyping strategies in depression genomics may apply to anxiety genomics.
Larger samples ascertained from brief phenotyping may increase loci discovery, but some
associations may not be disorder-specific. An improvement in power may lead to disorder-
specific biology or non-specific treatment targets of broader phenotypes such as neuroticism.
Genetic risk prediction may improve, but identify a broader group of individuals than the
specific disorder under analysis140. Therefore, both brief and detailed phenotyping are of
research value. Genetic datasets with both brief and detailed phenotyping are needed to make
significant progress in anxiety disorder genomics and ultimately translate GWAS findings for
clinical utility139.
Leveraging genomic data to understand pervasive comorbidity and clinical
heterogeneity
As discussed earlier, with evidence from twin studies, the pervasive comorbidity across
anxiety disorders and depression is likely partly driven by genetic overlap. Findings from
GWAS data further support this notion. GWAS consistently report substantial polygenic
overlap between anxiety and depression and other psychopathologies123,124,131,134. The largest
GWASs of anxiety and MDD show a genetic correlation of 0.72123,134. Estimates from other
GWAS are similar, with 0.78 reported between a clinically ascertained MDD phenotype and
lifetime anxiety disorder and up to 0.88 with GAD symptoms124,131. Neuroticism also shows
high genetic correlations with anxiety and depression (0.69-0.73)123,125,131,134, providing further
support for genetic influences on shared negative affectivity driving comorbidity. Significant
global genetic correlations have also been reported between anxiety and depression and a
range of other psychiatric disorders, health, and behavioural traits123,125,131,134. This broad array
of significant genetic correlations calculated from GWAS data can be leveraged for various
analyses to better understand shared and distinct aetiology106.
Unlike twin studies, molecular genetic assessment of disorder subfactors, such as the
distress-fear subdomain, is still in its infancy. A major limiting factor is the availability of
detailed phenotyping to measure subtypes for GWAS sufficiently. Anxiety and depression

Page 30
29
disorder subtype GWASs are needed to discern subtype-specific from transdiagnostic
common genetic influences. Failure to consider clinical heterogeneity may result in
overlooking specific biological pathways that can provide important insights into the genomic
influences on psychopathology. Furthermore, variable clinical heterogeneity across studies,
for example, inconsistent groupings of subtypes, may lead to inconsistent findings140.
More progress has been made in examining the genomic differences between the subtypes
of depression than for anxiety disorders. Subtyping of depression has found some evidence
that genetic heterogeneity reflects clinical heterogeneity143–145, although to a lesser extent in
some studies133,146,147. Differences in SNP-based heritability estimates have been reported.
More severe subtypes, including those treated with ECT, atypical, recurrent, and depression
with anxiety comorbidity, have shown higher heritability estimates than less clinically severe
forms of depression143,145. Genetic correlations across a range of subtypes from one study
suggested that 30-70% of genetic influences are shared143. Testing genetic differences
between subtypes of differing severity with a range of other complex traits has revealed
significant differences in genetic overlap with other psychiatric disorders, BMI, personality and
cognitive-related traits143,145,148. The more severe subtypes of depression are genetically less
similar to neuroticism145 and more similar to bipolar disorder and schizophrenia132. Such
analyses shed light on the genetic spectrum from manic to distress clusters (Figure 3),
showing both shared and unique genetic influences underlying depression and bipolar
disorder subtypes148. GWAS of depression subtypes have been largely underpowered for loci-
discovery and thus comparison149, but recent GWAS reveal evidence for some subtype-
specific loci143. Inconsistencies in subtyping have slowed progress in depression-subtype
genomics140.

Page 31
30
Figure 3: An illustration of the genomic continuum of the mood disorder spectrum
from mania genomics to internalising genomics.
Adapted from Coleman et al.148.
Examining anxiety disorder subtypes at the genome-wide level has been especially limited in
psychiatric genomics. This is even though diagnostic subtypes of anxiety disorders are more
well-established than depression subtypes5. Anxiety genomics has lagged behind due to the
lack of genetic datasets with detailed phenotyping on anxiety. Datasets with detailed
measures on all five anxiety disorders are even more scarce. Thus, a more prominent focus
has been placed on ‘any’ anxiety disorder groupings to maximise power122,131,134,135. It remains
unclear what genomic influences are shared or specific to individual anxiety disorders without
assessing comorbid conditions.
GWAS of individual anxiety disorders has been largely underpowered for loci-discovery and
genetic correlation analyses to assess disorder-specific and transdiagnostic genetic liability.
Recent progress has been made on GWAS of current GAD symptoms130,134, but well-powered
GWAS of lifetime GAD has been limited150. GWAS of individual fear-based disorders is further
lacking151. One genome-wide significant locus has been reported in a GWAS of panic
disorder152, and one in a GWAS of agoraphobia symptoms153. Therefore, a comprehensive
assessment of the distress-fear model at the molecular genetic level is lacking. Preliminary
GWAS findings in the UK Biobank indicate some genetic specificity among the fear-based
disorders that is not shared with distress disorders150. Chapter 2 expands upon this study by
conducting GWAS meta-analyses of fear-based disorders and GAD with newly established
Bipolar
disorder
type I
Major
depressive
disorder
Bipolar
disorder
type II
Mania
Internalising

Page 32
31
datasets that have detailed phenotyping on all five anxiety disorders. An improvement in
GWAS power enables a more comprehensive assessment of the common genetic variant
architecture of GAD and fear-based disorders and their genetic overlap with depression and
broad domains of other complex traits.
Social environmental influences on anxiety and depression
Research on the genetic basis of anxiety and depression is important for understanding
disorder aetiology, particularly for advancing our understanding of biology. However,
incorporating environmental information into genetic analyses is necessary to understand the
nature of genetic signals and the interplay between biological and social factors140. The
following sections of this chapter will first discuss how exposure to social environmental factors
is not independent of genetic influences. Psychological factors are discussed second, which
are the lens through which we experience environmental exposures and act as a bridge
between genetic and environmental influences on psychopathology. Figure 4 illustrates the
mechanisms involved in the interplay between genomics and different types of experiences in
the risk and treatment of common mental disorders.
Figure 4: An illustration of the potential mechanisms underpinning the course and
resolution of common mental disorders, which are influenced by the interplay
between genetic, socio-environmental and psychological factors.
Brown arrows represent genetic influences on biological pathways associated with the risk of
developing subtypes of common mental disorders, measured through genome-wide
association studies (GWAS). Experiences of negative and positive exposures and their
influence on the risk and treatment of psychopathology do not act independently of genetics.
Genomics can therefore be leveraged to understand these experiences. Blue arrows
represent the interplay between heritable characteristics associated with shaping adverse
environmental exposures and the risk of psychopathology, known as gene-environment
correlation. Green arrows represent the interaction between genetic liability to a disorder and
a psychological intervention (depicted here as a positive environmental exposure), leading to
variation in experiences and response, such as with psychological treatment.

Page 33
32
Approximately half of the phenotypic variability of anxiety and depressive disorders is not
explained by genetics. Twin studies (see Box 1) show that the remaining variance is primarily
attributed to the individual environment (sometimes called “non-shared”). The common
environment explains some variance during childhood but declines with age and is no longer
present during adulthood47,154. Individual-specific environmental liability is partially shared
across anxiety disorders and depression, though some disorder-specific influences are also
observed51. As such, unique individual-specific environmental factors are thought to explain
why anxiety disorders and depression present as distinct conditions155. Anxiety and
depression are associated with both positive and negative social environmental contexts.
Positive environments can mitigate the effects of adverse environmental risk factors. For
example, social support is a particularly important transdiagnostic buffer for the effects of
severe adverse environments, notably traumatic experiences156.
Psychologically traumatic experiences, which can be defined as those perceived by the
individual as devastating and overwhelming, are major risk factors for internalising disorders
and psychopathology more broadly5. Traumas experienced during childhood are the most
robust transdiagnostic environmental risk factors31,157. Childhood trauma is reported to
increase psychopathology risk by approximately two-fold158,159. The mechanisms underlying
the causal effects of trauma on psychopathology are an ongoing area of research. A recent
meta-analysis found that after controlling for pre-existing risk factors such as environmental
adversities and genetics, an attenuated but significant causal effect of childhood trauma on
broad psychopathology remained160. Transdiagnostic theories on the long-term biological
effects of early-life trauma include altered brain structure and function, such as disruption to
the development of the hypothalamic-pituitary-adrenal axis. This may lead to elevated
glucocorticoid signalling in response to stress and abnormal brain development with long-
lasting effects on emotion and behaviour161,162. Difficulties with emotions include dysregulation
and increased emotional reactivity, which, together with elevated informational processing of
social cues as threatening, can influence maladaptive behavioural responses156. Given its
transdiagnostic effects, understanding the risk and consequences of trauma is an important
area of research. Furthermore, targeting trauma exposure in prevention and intervention
strategies may be more effective in reducing psychopathology risk than disorder-specific
environmental risk factors157.
The heritability of environmental measures
A complexity of disentangling the causal effects of environmental exposures on
psychopathology is that the environment does not operate independently of genetics. Twin
studies show that almost all measures of the environment are themselves genetically

Page 34
33
influenced163. This includes the reporting of positive, negative, and traumatic social exposures,
which have a heritability ranging from 24-62%163–167. Significant SNP-based heritability
estimates have also been found for reported traumatic and stressful exposures (6-
30%)132,168,169. Twin studies find that life events dependent on one’s behaviour are more
heritable than independent events163,170,171, likely due to heritable behavioural characteristics
influencing the environments we are exposed to. These environments can be shaped
passively by our parents during childhood, or we actively select them in later life and by how
others engage with us. Correlations between genetic factors and environmental exposures
can arise through these mechanisms, termed gene-environment correlation172,173. As such,
the association between environmental measures and psychopathology may partly be
explained by shared genetic influences on both the likelihood of environmental exposure and
the development of psychopathology.
Identifying the genetically influenced traits that contribute to the heritability of environmental
measures would elucidate key traits involved in gene-environment correlations. Such findings
help narrow down characteristics to target in follow-up analyses assessing causal pathways
and ultimately inform intervention or prevention strategies. Multivariate twin modelling has
been used to deconstruct the heritability of an environmental measure by jointly modelling
genetically correlated personality and behavioural traits174,175. For example, one study found
that genetic propensity for non-cognitive traits is as important as genetic influences on
cognitive ability in explaining the heritability of educational attainment. This study highlighted
plausible alternative candidates for non-cognitive interventions to improve engagement with
the schooling environment174. As noted earlier, recent advancements in genomics research
enable the systematic assessment and multi-trait modelling of genetic correlations between a
broad range of traits, including environmental exposures and psychiatric and behavioural
traits115. For example, genomic multiple regression models in Genomic SEM have been used
to estimate genetic correlations between psychiatric disorders and smoking behaviours,
independent of other heritable behavioural traits176. This method could also be used to
deconstruct the SNP-based heritability of adverse environments and identify uniquely
genetically associated psychiatric and behavioural traits. Thus, such approaches provide the
opportunity to better understand key environmental risk factors for psychopathology at the
genomic level. Chapter 3 explores the genomic influences of multiple traits on vulnerability to
childhood trauma and aims to identify the traits that may be involved in gene-environment
correlation mechanisms through genomic multiple regression modelling.

Page 35
34
Heritable psychological influences on anxiety and depression: from risk to
treatment
As outlined earlier, twin studies show that the individual environment explains more
phenotypic variation in anxiety and depression than the common environment. This is partly
due to the important role of individual differences in psychological influences that shape how
people experience their environment, which is unique to each person177,178. Genetics also
plays a role in how sensitive we are and the context in which we perceive exposures, which
in turn influences our response179. Genetics can therefore be leveraged to understand
characteristics associated with vulnerability to experiencing adverse exposures as traumatic
and a greater risk of subsequent psychopathology180.
Heritable influences on subjective self-reports of adverse experiences
Various genetically influenced psychological and cognitive factors impact sensitivity to
adverse environments. The stress-diathesis model describes sensitivity to negative (i.e.
stressful) environmental experiences and explains why not all those exposed to adverse
environments develop a common mental health problem. Genetic pathogenic effects depend
on environmental exposure and vice versa13,179,181,182. It is plausible that psychological factors
that influence sensitivity to adverse environments also influence reporting of events as
stressful or traumatic. As such, using self-reports to capture environmental exposures is often
conceptualised as a limitation to studies due to potential biases in reporting183. Meta-analyses
show that subjective and objective measures of adversity identify largely different groups of
individuals184. Thus, if research aims only to understand the influences on objective exposure,
then subjective reporting may capture influences on reporting biases as opposed to
mechanisms associated with exposure. Such research is important for prevention strategies.
However, understanding the genetic components contributing to subjective reporting is also
valuable. The subjective experience of trauma, as captured by retrospective self-reports, is
key for developing psychopathology, more so than trauma exposure. Some individuals
identified as exposed through objective measures, such as court documentation of childhood
maltreatment, do not subjectively report trauma in later life. These individuals are less likely
to develop post-traumatic psychopathology than those who retrospectively self-report
trauma184. This may reflect a group of individuals with low sensitivity to adverse environments,
which acts as a protective factor and resilience to adversity. Thus, understanding the
psychological and cognitive factors associated with retrospective and subjective reporting may
be informative for psychopathology intervention.

Page 36
35
Psychological factors, including personality traits and cognitive biases, may influence how
individuals experience and engage with adverse environments and later risk of
psychopathology. Neuroticism is associated with cognitive biases towards negative stimuli,
elevated stress levels, and environmental sensitivity185,186. Twin studies suggest that genetics
primarily explain the correlation between neuroticism and adverse environmental sensitivity186,
highlighting the role of genetics in the subjective appraisal of adversity. Individuals with higher
levels of neuroticism tend to self-report more adversity in the absence of prospective records.
This is in contrast to agreeableness, which is associated with lower rates of retrospective
reports than indicated by the level of prospective accounts187. Higher neuroticism is also linked
with reporting traumatic and stressful events as more central to one's identity, a risk factor for
later psychopathology188,189. A potential mechanism explaining the link between neuroticism
and posttraumatic psychopathology is the increased emotional availability of trauma memories
through more frequent retrieval and rehearsal and, thus, maintenance of the memory189. A
genetic overlap has been observed between neuroticism, depressive symptoms, and
perceived stress190. Thus, genetic influences on neuroticism may influence the reporting of
events as traumatic166. Indeed, genetic correlations between GWASs of neuroticism and
retrospective self-reports of trauma have been observed132. Chapter 3 explores the extent to
which genomic influences on psychological traits such as neuroticism can explain the
heritability of retrospectively self-reporting adverse environments, aiming to identify key traits
involved in the experience of adverse environments.
Heritable influences on positive experiences and application to psychological
treatment
Genetic influences on psychological factors impact not only vulnerability to respond negatively
following trauma (as in the diathesis-stress model) but also sensitivity to positive
environments. The differential susceptibility model extends environmental sensitivity to
genetic influences on psychological factors affecting the experience and response to both
negative and positive environments. Under this framework, we can leverage genetic
information to understand who is more likely to benefit from support environments, providing
a more positive outlook on intervention research of psychopathology. Indeed, a core focus on
anxiety and depression genomics is understanding risk factors. However, genomics can also
be harnessed to improve treatment selection. For example, individuals with high
environmental sensitivity may translate to a better response to protective and supportive
exposures, such as psychological intervention179,191,192.
There is substantial individual variability observed in treatment response for anxiety and
depression. Given the overlap in risk factors, it is not surprising that treatments for anxiety and

Page 37
36
depression are highly similar, with comparable treatment response rates across disorders31.
Medication and psychological therapy are both first-line treatments for anxiety and depression.
However, not all individuals respond to them equally, with approximately only 50% responding
to current treatments193,194. Predicting who will respond best to which treatment is poor and is
therefore often prescribed in a trial and error fashion13,194. Research understanding the factors
that influence treatment response is needed to aid in identifying suitable predictors to guide
treatment selection.
Genetic influences on antidepressant response show the potential for using genetic
information in guiding treatment. Psychiatric pharmacogenomics, which aims to harness
genetic variants associated with variability in response to medication to guide treatment
selection, has grown over recent years with some progress in antidepressant response
research. Although findings have been mixed, a meta-analysis of randomised controlled trials
of pharmacogenomic testing for antidepressant response showed a 71% increased likelihood
of depressive symptom remission in individuals receiving pharmaco-guided antidepressant
prescribing compared to standard practice prescribing195. Based on individual-level estimates,
the SNP-based heritability of antidepressant response is approximately 42%196 and 13% for
depressive symptom remission following antidepressant treatment197. These findings
represent considerable progress in the field of antidepressant response genetics. However,
antidepressants are not always beneficial to patients, and an integrated approach with
psychological therapy and medication outperforms pharmacotherapy alone. Thus, a combined
understanding of factors associated with response to both forms of treatment would be of even
greater clinical utility in guiding treatment selection.
Identifying predictors of improvement in anxiety and depressive symptoms following
psychological therapy is an ongoing area of research. Cognitive behavioural therapy (CBT) is
the gold standard psychological treatment for anxiety and depression198, including disorder-
specific and transdiagnostic approaches199, which are equally as effective200,201. Grouping
internalising disorders may prove useful in improving the prediction of psychological treatment
outcomes31. The ultimate goal is to incorporate a range of variables into predictive models to
aid in treatment decision-making202. Adding genetic factors to these models requires
advancements in examining the genetics of psychological treatment outcomes.
Considering there are genetic influences on a range of behavioural, psychological and
cognitive traits, which in turn influence response to positive environments163, there is likely a
genetic component to psychological treatment. No twin studies have found a significant
heritability of outcomes following psychological therapy, also known as ‘therapygenetics’.
However, evidence from examining related phenotypes suggests that a heritable component

Page 38
37
exists. Twin studies report a significant heritability of fear extinction of 15-36%, a key
mechanism involved in exposure-based CBT for anxiety disorders203,204. Half of the genetic
influences on fear acquisition also overlap with fear extinction. This suggests that some
genetic influences on anxiety-related traits could be used to understand responses to
psychological therapy203. For example, polygenic scores from existing well-powered GWAS of
anxiety or depression may be useful in predicting psychological therapy outcomes alongside
other predictors. As such, incorporating pre-existing polygenic scores from well-powered
GWAS of related complex traits may be a promising avenue for therapygenetics research205.
Therapygenetics can be understood through the framework of gene-environment interaction,
in which psychological intervention is the environmental exposure and genetics influence
sensitivity to the exposure206. Preliminary findings using polygenic scores derived from a small
GWAS of environmental sensitivity in twins indicates that a higher genetic propensity for
environmental sensitivity is associated with a better response to more intensive CBT than less
intensive207. As was the case for many polygenic traits in candidate gene studies, early
attempts to identify specific genetic variants with robust gene-environment interaction effects
on psychological treatment outcomes were unsuccessful206,208. Response to psychological
therapy is unlikely to be influenced by a few genes, but rather highly polygenic, as is the case
for all heritable psychological traits.
A challenge for genome-wide therapygenetics is phenotyping at the scale required for GWAS.
GWAS of prognostic outcomes following CBT for anxiety and depression have been limited in
power due to the difficulty in ascertaining such phenotypes. The largest published GWAS
meta-analysis thus far was small, with 2,724 participants, and did not detect a significant SNP-
based heritability or genome-wide significant loci209. Alternative phenotyping strategies are
required to achieve sufficient power to detect the thousands of loci underpinning complex
traits. As with anxiety and depressive disorder genomics research, an improvement in power
has largely been achieved through brief phenotyping. The utility of brief phenotyping in GWAS
of psychological therapy outcomes has yet to be explored. Furthermore, subjective self-
reports of benefitting from psychological therapy may be useful in capturing genetic influences
that impact the perception of experiences, as with self-reports of environmental adversity.
Chapter 4 uses brief self-reports of psychological treatment outcomes to increase the sample
size for GWAS and prediction models that incorporate polygenic scores of related traits.

Page 39
38
Summary and aims
The overarching aim of this thesis was to further the understanding of the risk and treatment
of common mental disorders by integrating bio-psycho-social components and leveraging
large genomic datasets (Figure 1). Chapters 2 and 3 focus on understanding genetic
influences on risk factors, while Chapter 4 explores genetic influences on treatment outcomes
(see Figure 4). Chapter 2 focuses more on the biological and genomic mechanisms
associated with clinical heterogeneity, whilst Chapters 3 and 4 harness genomic data to
understand psychological influences on environmental experiences. To improve statistical
power for genomic analyses of phenotypes that currently lack GWAS with sufficient sample
sizes, Chapters 2 and 4 leverage novel genotyped datasets with detailed measures of anxiety
and depression outcomes. All three empirical chapters harness pre-existing, well-powered
GWAS summary statistics across bio-psycho-social domains of complex traits related to
common mental disorders. This includes psychopathologies and physical health problems
commonly co-occurring with anxiety and depression, reported socio-environmental
experiences, and cognitive-related and psychological traits. Examining a broad range of
complex traits enables the assessment of shared and distinct genetic influences on common
mental disorders and the interplay with psychosocial influences at a large scale.

Page 40
39
References
1. Organization, W. H. & Others. Depression and other common mental disorders:
global health estimates.
https://apps.who.int/iris/bitstream/handle/10665/254610/WHO-MSD-MER-2017.2-
eng.pdf (2017).
2. Kessler, R. C. et al. The global burden of mental disorders: an update from the
WHO World Mental Health (WMH) surveys. Epidemiol. Psichiatr. Soc. 18, 23–33
(2009).
3. Lim, G. Y. et al. Prevalence of Depression in the Community from 30 Countries
between 1994 and 2014. Sci. Rep. 8, 2861 (2018).
4. Somers, J. M., Goldner, E. M., Waraich, P. & Hsu, L. Prevalence and incidence
studies of anxiety disorders: a systematic review of the literature. Can. J. Psychiatry
51, 100–113 (2006).
5. American Psychiatric Association. Diagnostic and Statistical Manual of Mental
Disorders (DSM-5�). (American Psychiatric Pub, 2013).
6. Aquino, J. P. D., De Aquino, J. P., Londono, A. & Carvalho, A. F. An Update on the
Epidemiology of Major Depressive Disorder Across Cultures. Understanding
Depression 309–315 Preprint at https://doi.org/10.1007/978-981-10-6580-4_25
(2018).
7. Breslau, J., Lane, M., Sampson, N. & Kessler, R. C. Mental disorders and
subsequent educational attainment in a US national sample. J. Psychiatr. Res. 42,
708–716 (2008).
8. Fergusson, D. M. & Woodward, L. J. Mental health, educational, and social role
outcomes of adolescents with depression. Arch. Gen. Psychiatry 59, 225–231
(2002).
9. McDaid, D., Park, A. L., Davidson, G., John, A. & Knifton, L. The economic case for
investing in the prevention of mental health conditions in the UK. (2022).
10. Craske, M. G. & Stein, M. B. Anxiety. Focus 15, 199–210 (2017).
11. Malhi, G. S. & John Mann, J. Depression. The Lancet vol. 392 2299–2312 Preprint
at https://doi.org/10.1016/s0140-6736(18)31948-2 (2018).
12. Too, L. S. et al. The association between mental disorders and suicide: A systematic
review and meta-analysis of record linkage studies. J. Affect. Disord. 259, 302–313
(2019).
13. Craske, M. G. et al. Anxiety disorders. Nat Rev Dis Primers 3, 17024 (2017).
14. Harald, B. & Gordon, P. Meta-review of depressive subtyping models. J. Affect.
Disord. 139, 126–140 (2012).
15. Toft, T. et al. Mental disorders in primary care: prevalence and co-morbidity among
disorders. results from the functional illness in primary care (FIP) study. Psychol.
Med. 35, 1175–1184 (2005).
16. Kessler, R. C., Merikangas, K. R. & Wang, P. S. Prevalence, comorbidity, and
service utilization for mood disorders in the United States at the beginning of the
twenty-first century. Annu. Rev. Clin. Psychol. 3, 137–158 (2007).
17. Kessler, R. C. et al. Comorbidity of DSM–III–R Major Depressive Disorder in the
General Population: Results from the US National Comorbidity Survey. Br. J.
Psychiatry 168, 17–30 (1996).

Page 41
40
18. Kessler, R. C. et al. The epidemiology of major depressive disorder: results from the
National Comorbidity Survey Replication (NCS-R). JAMA 289, 3095–3105 (2003).
19. Kessler, R. C. et al. Lifetime prevalence and age-of-onset distributions of DSM-IV
disorders in the National Comorbidity Survey Replication. Arch. Gen. Psychiatry 62,
593–602 (2005).
20. Hirschfeld, R. M. A. The Comorbidity of Major Depression and Anxiety Disorders:
Recognition and Management in Primary Care. Prim. Care Companion J. Clin.
Psychiatry 3, 244–254 (2001).
21. Lamers, F. et al. Comorbidity patterns of anxiety and depressive disorders in a large
cohort study: the Netherlands Study of Depression and Anxiety (NESDA). J. Clin.
Psychiatry 72, 341–348 (2011).
22. Steffen, A., N�bel, J., Jacobi, F., B�tzing, J. & Holstiege, J. Mental and somatic
comorbidity of depression: a comprehensive cross-sectional analysis of 202
diagnosis groups using German nationwide ambulatory claims data. BMC
Psychiatry 20, 142 (2020).
23. Penninx, B. W., Pine, D. S., Holmes, E. A. & Reif, A. Anxiety disorders. Lancet 397,
914–927 (2021).
24. Prince, M. et al. No health without mental health. Lancet 370, 859–877 (2007).
25. Kraus, C., Kadriu, B., Lanzenberger, R., Zarate, C. A., Jr & Kasper, S. Prognosis
and Improved Outcomes in Major Depression: A Review. Focus 18, 220–235
(2020).
26. Noyes, R., Jr. Comorbidity in generalized anxiety disorder. Psychiatr. Clin. North
Am. 24, 41–55 (2001).
27. Kalin, N. H. The Critical Relationship Between Anxiety and Depression. Am. J.
Psychiatry 177, 365–367 (2020).
28. Clark, L. A. & Watson, D. Distress and fear disorders: an alternative empirically
based taxonomy of the ‘mood’ and ‘anxiety’ disorders. Br. J. Psychiatry 189, 481–
483 (2006).
29. Eysenck, M. W. & Fajkowska, M. Anxiety and depression: toward overlapping and
distinctive features. Cogn. Emot. 32, 1391–1400 (2018).
30. Crocq, M.-A. The history of generalized anxiety disorder as a diagnostic category.
Dialogues Clin. Neurosci. 19, 107–116 (2017).
31. Watson, D. et al. Validity and utility of Hierarchical Taxonomy of Psychopathology
(HiTOP): III. Emotional dysfunction superspectrum. World Psychiatry 21, 26–54
(2022).
32. Conway, C. C., Forbes, M. K., South, S. C. & HiTOP Consortium. A Hierarchical
Taxonomy of Psychopathology (HiTOP) Primer for Mental Health Researchers. Clin.
Psychol. Sci. 10, 236–258 (2022).
33. Krueger, R. F. et al. Validity and utility of Hierarchical Taxonomy of Psychopathology
(HiTOP): II. Externalizing superspectrum. World Psychiatry vol. 20 171–193 Preprint
at https://doi.org/10.1002/wps.20844 (2021).
34. Kotov, R. et al. Validity and utility of Hierarchical Taxonomy of Psychopathology
(HiTOP): I. Psychosis superspectrum. World Psychiatry 19, 151–172 (2020).
35. Stanton, K. et al. An Initial Investigation of the Joint Classification of Hypomania-
and Neurodevelopmental Disorder-Relevant Dimensions Within the Hierarchical
Taxonomy of Psychopathology. Assessment 30, 414–432 (2023).
36. Stanton, K., DeLucia, E. A., Brown, M. F. D. & McDonnell, C. G. Advancing
understanding of the classification of broad autism phenotype and attention-

Page 42
41
deficit/hyperactivity disorder symptom dimensions within the Hierarchical Taxonomy
of Psychopathology. Personal. Ment. Health 15, 113–123 (2021).
37. Caspi, A. et al. The p Factor: One General Psychopathology Factor in the Structure
of Psychiatric Disorders? Clin. Psychol. Sci. 2, 119–137 (2014).
38. Watson, D. Rethinking the mood and anxiety disorders: a quantitative hierarchical
model for DSM-V. J. Abnorm. Psychol. 114, 522–536 (2005).
39. Kendler, K. S. The structure of psychiatric science. Am. J. Psychiatry 171, 931–938
(2014).
40. Hettema, J. M., Neale, M. C. & Kendler, K. S. A review and meta-analysis of the
genetic epidemiology of anxiety disorders. Am. J. Psychiatry 158, 1568–1578
(2001).
41. Sullivan, P. F., Neale, M. C. & Kendler, K. S. Genetic epidemiology of major
depression: review and meta-analysis. Am. J. Psychiatry 157, 1552–1562 (2000).
42. Smoller, J. W. The Genetics of Stress-Related Disorders: PTSD, Depression, and
Anxiety Disorders. Neuropsychopharmacology 41, 297–319 (2016).
43. Polderman, T. J. C. et al. Meta-analysis of the heritability of human traits based on
fifty years of twin studies. Nat. Genet. 47, 702–709 (2015).
44. Craddock, N., O’Donovan, M. C. & Owen, M. J. The genetics of schizophrenia and
bipolar disorder: dissecting psychosis. J. Med. Genet. 42, 193–204 (2005).
45. Kendall, K. M. et al. The genetic basis of major depression. Psychol. Med. 51,
2217–2230 (2021).
46. Rice, F. Genetics of childhood and adolescent depression: insights into etiological
heterogeneity and challenges for future genomic research. Genome Med. 2, 68
(2010).
47. Nivard, M. G. et al. Stability in symptoms of anxiety and depression as a function of
genotype and environment: a longitudinal twin study from ages 3 to 63 years.
Psychol. Med. 45, 1039–1049 (2015).
48. Hannigan, L. J., Walaker, N., Waszczuk, M. A., McAdams, T. A. & Eley, T. C.
Aetiological influences on stability and change in emotional and behavioural
problems across development: a systematic review. Psychopathol Rev 4, 52–108
(2017).
49. Waszczuk, M. A., Zavos, H. M. S., Gregory, A. M. & Eley, T. C. The stability and
change of etiological influences on depression, anxiety symptoms and their co-
occurrence across adolescence and young adulthood. Psychol. Med. 46, 161–175
(2016).
50. Cerd�, M., Sagdeo, A., Johnson, J. & Galea, S. Genetic and environmental
influences on psychiatric comorbidity: a systematic review. J. Affect. Disord. 126,
14–38 (2010).
51. Waszczuk, M. A., Zavos, H. M. S., Gregory, A. M. & Eley, T. C. The phenotypic and
genetic structure of depression and anxiety disorder symptoms in childhood,
adolescence, and young adulthood. JAMA Psychiatry 71, 905–916 (2014).
52. Middeldorp, C. M., Cath, D. C., Van Dyck, R. & Boomsma, D. I. The co-morbidity of
anxiety and depression in the perspective of genetic epidemiology. A review of twin
and family studies. Psychol. Med. 35, 611–624 (2005).
53. Mosing, M. A. et al. Genetic and environmental influences on the co-morbidity
between depression, panic disorder, agoraphobia, and social phobia: a twin study.
Depress. Anxiety 26, 1004–1011 (2009).
54. Hettema, J. M. et al. Genetic and environmental risk structure of internalizing

Page 43
42
psychopathology in youth. Depress. Anxiety 37, 540–548 (2020).
55. Kendler, K. S. et al. The structure of genetic and environmental risk factors for
syndromal and subsyndromal common DSM-IV axis I and all axis II disorders. Am.
J. Psychiatry 168, 29–39 (2011).
56. Hettema, J. M., Neale, M. C., Myers, J. M., Prescott, C. A. & Kendler, K. S. A
population-based twin study of the relationship between neuroticism and
internalizing disorders. Am. J. Psychiatry 163, 857–864 (2006).
57. Tackett, J. L. et al. Common genetic influences on negative emotionality and a
general psychopathology factor in childhood and adolescence. J. Abnorm. Psychol.
122, 1142–1153 (2013).
58. Pettersson, E., Larsson, H. & Lichtenstein, P. Common psychiatric disorders share
the same genetic origin: a multivariate sibling study of the Swedish population. Mol.
Psychiatry 21, 717–721 (2016).
59. Kendler, K. S. & Myers, J. The boundaries of the internalizing and externalizing
genetic spectra in men and women. Psychol. Med. 44, 647–655 (2014).
60. Hettema, J. M., Prescott, C. A., Myers, J. M., Neale, M. C. & Kendler, K. S. The
structure of genetic and environmental risk factors for anxiety disorders in men and
women. Arch. Gen. Psychiatry 62, 182–189 (2005).
61. Kendler, K. S., Prescott, C. A., Myers, J. & Neale, M. C. The structure of genetic and
environmental risk factors for common psychiatric and substance use disorders in
men and women. Arch. Gen. Psychiatry 60, 929–937 (2003).
62. Visscher, P. M., Hill, W. G. & Wray, N. R. Heritability in the genomics era--concepts
and misconceptions. Nat. Rev. Genet. 9, 255–266 (2008).
63. Wray, N. & Visscher, P. Estimating trait heritability. Nature education.
64. Barry, C.-J. S. et al. How to estimate heritability: a guide for genetic epidemiologists.
Int. J. Epidemiol. (2022) doi:10.1093/ije/dyac224.
65. Mayhew, A. J. & Meyre, D. Assessing the Heritability of Complex Traits in Humans:
Methodological Challenges and Opportunities. Curr. Genomics 18, 332–340 (2017).
66. Hill, W. G., Goddard, M. E. & Visscher, P. M. Data and theory point to mainly
additive genetic variance for complex traits. PLoS Genet. 4, e1000008 (2008).
67. Yang, J., Zeng, J., Goddard, M. E., Wray, N. R. & Visscher, P. M. Concepts,
estimation and interpretation of SNP-based heritability. Nat. Genet. 49, 1304–1310
(2017).
68. Yang, J. et al. Common SNPs explain a large proportion of the heritability for human
height. Nat. Genet. 42, 565–569 (2010).
69. Yang, J., Lee, S. H., Goddard, M. E. & Visscher, P. M. GCTA: a tool for genome-
wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).
70. Baselmans, B. M. L., Yengo, L., van Rheenen, W. & Wray, N. R. Risk in Relatives,
Heritability, SNP-Based Heritability, and Genetic Correlations in Psychiatric
Disorders: A Review. Biol. Psychiatry 89, 11–19 (2021).
71. Bulik-Sullivan, B. K. et al. LD Score regression distinguishes confounding from
polygenicity in genome-wide association studies. Nature Genetics vol. 47 291–295
Preprint at https://doi.org/10.1038/ng.3211 (2015).
72. Lee, S. H., Wray, N. R., Goddard, M. E. & Visscher, P. M. Estimating missing
heritability for disease from genome-wide association studies. Am. J. Hum. Genet.
88, 294–305 (2011).
73. Andreassen, O. A., Hindley, G. F. L., Frei, O. & Smeland, O. B. New insights from
the last decade of research in psychiatric genetics: discoveries, challenges and

Page 44
43
clinical implications. World Psychiatry 22, 4–24 (2023).
74. Roses, A. D. The genome era begins. Nat. Genet. 33, 217–217 (2003).
75. Ikegawa, S. A short history of the genome-wide association study: where we were
and where we are going. Genomics Inform. 10, 220–225 (2012).
76. Hofker, M. H., Fu, J. & Wijmenga, C. The genome revolution and its role in
understanding complex diseases. Biochim. Biophys. Acta 1842, 1889–1895 (2014).
77. Michaelson, J. J. Genetic Approaches to Understanding Psychiatric Disease.
Neurotherapeutics 14, 564–581 (2017).
78. Edwards, S. L., Beesley, J., French, J. D. & Dunning, A. M. Beyond GWASs:
illuminating the dark road from association to function. Am. J. Hum. Genet. 93, 779–
797 (2013).
79. Knopik, V. S., Neiderhiser, J. M., DeFries, J. C. & Plomin, R. Behavioral Genetics.
(Macmillan Learning, 2016).
80. Visscher, P. M., Yengo, L., Cox, N. J. & Wray, N. R. Discovery and implications of
polygenicity of common diseases. Science 373, 1468–1473 (2021).
81. Karki, R., Pandya, D., Elston, R. C. & Ferlini, C. Defining ‘mutation’ and
‘polymorphism’ in the era of personal genomics. BMC Med. Genomics 8, 37 (2015).
82. Brookes, A. J. The essence of SNPs. Gene 234, 177–186 (1999).
83. Cavalli-Sforza, L. L. & Bodmer, W. F. The Genetics of Human Populations. (Courier
Corporation, 1999).
84. Arias, T. D., Jorge, L. F. & Barrantes, R. Uses and misuses of definitions of genetic
polymorphism. A perspective from population pharmacogenetics. Br. J. Clin.
Pharmacol. 31, 117–119 (1991).
85. Day, I. N. M. dbSNP in the detail and copy number complexities. Hum. Mutat. 31, 2–
4 (2010).
86. Eichler, E. E. Copy number variation and human disease. Nat Educ.
87. 1000 Genomes Project Consortium et al. An integrated map of genetic variation
from 1,092 human genomes. Nature 491, 56–65 (2012).
88. Consortium, †the International Hapmap & †The International HapMap Consortium.
The International HapMap Project. Nature vol. 426 789–796 Preprint at
https://doi.org/10.1038/nature02168 (2003).
89. Zhao, S., Jiang, L., Yu, H. & Guo, Y. GTQC: Automated Genotyping Array Quality
Control and Report. J Genomics 10, 39–44 (2022).
90. Visscher, P. M. et al. 10 Years of GWAS Discovery: Biology, Function, and
Translation. Am. J. Hum. Genet. 101, 5–22 (2017).
91. Slatkin, M. Linkage disequilibrium--understanding the evolutionary past and
mapping the medical future. Nat. Rev. Genet. 9, 477–485 (2008).
92. Schaid, D. J., Chen, W. & Larson, N. B. From genome-wide associations to
candidate causal variants by statistical fine-mapping. Nat. Rev. Genet. 19, 491–504
(2018).
93. Cano-Gamez, E. & Trynka, G. From GWAS to Function: Using Functional Genomics
to Identify the Mechanisms Underlying Complex Diseases. Frontiers in Genetics vol.
11 Preprint at https://doi.org/10.3389/fgene.2020.00424 (2020).
94. Watanabe, K., Taskesen, E., van Bochoven, A. & Posthuma, D. Functional mapping
and annotation of genetic associations with FUMA. Nat. Commun. 8, 1826 (2017).
95. Peterson, R. E. et al. Genome-wide Association Studies in Ancestrally Diverse
Populations: Opportunities, Methods, Pitfalls, and Recommendations. Cell 179,
589–603 (2019).

Page 45
44
96. Reich, D. E. et al. Linkage disequilibrium in the human genome. Nature 411, 199–
204 (2001).
97. Uffelmann, E. et al. Genome-wide association studies. Nature Reviews Methods
Primers 1, 1–21 (2021).
98. National Academies of Sciences, Engineering & Medicine. Using Population
Descriptors in Genetics and Genomics Research: A New Framework for an Evolving
Field. (2023).
99. Price, A. L. et al. Principal components analysis corrects for stratification in genome-
wide association studies. Nat. Genet. 38, 904–909 (2006).
100. Novembre, J. & Stephens, M. Interpreting principal component analyses of spatial
population genetic variation. Nat. Genet. 40, 646–649 (2008).
101. Visscher, P. M., Goddard, M. E., Derks, E. M. & Wray, N. R. Evidence-based
psychiatric genetics, AKA the false dichotomy between common and rare variant
hypotheses. Mol. Psychiatry 17, 474–485 (2012).
102. Fabbri, C. Genetics in psychiatry: Methods, clinical applications and future
perspectives. PCN Rep. 1, (2022).
103. Uffelmann, E. & Posthuma, D. Emerging Methods and Resources for Biological
Interrogation of Neuropsychiatric Polygenic Signal. Biol. Psychiatry 89, 41–53
(2021).
104. Wray, N. R. et al. Genome-wide association study of major depressive disorder: new
results, meta-analysis, and lessons learned. Mol. Psychiatry 17, 36–48 (2012).
105. Bulik-Sullivan, B. et al. An atlas of genetic correlations across human diseases and
traits. Nat. Genet. 47, 1236–1241 (2015).
106. van Rheenen, W., Peyrot, W. J., Schork, A. J., Lee, S. H. & Wray, N. R. Genetic
correlations of polygenic disease traits: from theory to practice. Nat. Rev. Genet.
(2019) doi:10.1038/s41576-019-0137-z.
107. Abdellaoui, A. & Verweij, K. J. H. Dissecting polygenic signals from genome-wide
association studies on human behaviour. Nat Hum Behav 5, 686–694 (2021).
108. Avinun, R. The E Is in the G: Gene-Environment-Trait Correlations and Findings
From Genome-Wide Association Studies. Perspect. Psychol. Sci. 15, 81–89 (2020).
109. Solovieff, N., Cotsapas, C., Lee, P. H., Purcell, S. M. & Smoller, J. W. Pleiotropy in
complex traits: challenges and strategies. Nat. Rev. Genet. 14, 483–495 (2013).
110. Zheng, J. et al. LD Hub: a centralized 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 vol. 33 272–279 Preprint
at https://doi.org/10.1093/bioinformatics/btw613 (2017).
111. Ronald, A. & Pain, O. A Revolution Is Brewing in How We Understand the Shared
Genetic Causes of Psychiatric Disorders. The American journal of psychiatry vol.
179 791–793 (2022).
112. Visscher, P. M. & Yang, J. A plethora of pleiotropy across complex traits. Nature
genetics vol. 48 707–708 (2016).
113. Krapohl, E. et al. Multi-polygenic score approach to trait prediction. Mol. Psychiatry
23, 1368–1374 (2018).
114. Grotzinger, A. D. Shared genetic architecture across psychiatric disorders. Psychol.
Med. 51, 2210–2216 (2021).
115. Grotzinger, A. D. et al. Genomic structural equation modelling provides insights into
the multivariate genetic architecture of complex traits. Nat Hum Behav 3, 513–525
(2019).

Page 46
45
116. Allegrini, A. G. et al. Multivariable G-E interplay in the prediction of educational
achievement. PLoS Genet. 16, e1009153 (2020).
117. Choi, S. W., Mak, T. S.-H. & O’Reilly, P. F. Tutorial: a guide to performing polygenic
risk score analyses. Nat. Protoc. 15, 2759–2772 (2020).
118. Trubetskoy, V. et al. Mapping genomic loci implicates genes and synaptic biology in
schizophrenia. Nature 604, 502–508 (2022).
119. Lewis, C. M. & Vassos, E. Polygenic risk scores: from research tools to clinical
instruments. Genome Med. 12, 44 (2020).
120. Psychiatric GWAS Consortium Coordinating Committee et al. Genomewide
association studies: history, rationale, and prospects for psychiatric disorders. Am.
J. Psychiatry 166, 540–556 (2009).
121. Sullivan, P. F. & Kendler, K. S. The state of the science in psychiatric genomics.
Psychol. Med. 51, 2145–2147 (2021).
122. Ask, H. et al. Genetic contributions to anxiety disorders: where we are and where we
are heading. Psychol. Med. 51, 2231–2246 (2021).
123. Levey, D. F. et al. Bi-ancestral depression GWAS in the Million Veteran Program
and meta-analysis in >1.2 million individuals highlight new therapeutic directions.
Nat. Neurosci. 24, 954–963 (2021).
124. Wray, N. R. et al. Genome-wide association analyses identify 44 risk variants and
refine the genetic architecture of major depression. Nat. Genet. 50, 668–681 (2018).
125. Howard, D. M. et al. Genome-wide meta-analysis of depression identifies 102
independent variants and highlights the importance of the prefrontal brain regions.
Nat. Neurosci. 22, 343–352 (2019).
126. Tubbs, J. D., Ding, J., Baum, L. & Sham, P. C. Immune dysregulation in depression:
Evidence from genome-wide association. Brain Behav Immun Health 7, 100108
(2020).
127. Smoller, J. W. Anxiety Genetics Goes Genomic. The American journal of psychiatry
vol. 177 190–194 (2020).
128. Smoller, J. W. Who’s afraid of anxiety genetics? Biological psychiatry vol. 69 506–
507 (2011).
129. Levey, D. F., Gelernter, J. & Polimanti, R. Reproducible genetic risk loci for anxiety:
results from∼ 200,000 participants in the Million Veteran Program. American Journal
of (2020).
130. Wendt, F. R. et al. Using phenotype risk scores to enhance gene discovery for
generalized anxiety disorder and posttraumatic stress disorder. Mol. Psychiatry
(2022) doi:10.1038/s41380-022-01469-y.
131. Purves, K. L. et al. A major role for common genetic variation in anxiety disorders.
Mol. Psychiatry 25, 3292–3303 (2020).
132. Coleman, J. R. I. et al. Genome-wide gene-environment analyses of major
depressive disorder and reported lifetime traumatic experiences in UK Biobank. Mol.
Psychiatry (2020) doi:10.1038/s41380-019-0546-6.
133. Hall, L. S. et al. Genome-wide meta-analyses of stratified depression in Generation
Scotland and UK Biobank. Transl. Psychiatry 8, 9 (2018).
134. Levey, D. F. et al. Reproducible Genetic Risk Loci for Anxiety: Results From
∼200,000 Participants in the Million Veteran Program. Am. J. Psychiatry 177, 223–
232 (2020).
135. Otowa, T. et al. Meta-analysis of genome-wide association studies of anxiety
disorders. Mol. Psychiatry 21, 1391–1399 (2016).

Page 47
46
136. Nagel, M. et al. Meta-analysis of genome-wide association studies for neuroticism in
449,484 individuals identifies novel genetic loci and pathways. Nat. Genet. 50, 920–
927 (2018).
137. Grotzinger, A. D., Fuente, J. de la, Priv�, F., Nivard, M. G. & Tucker-Drob, E. M.
Pervasive Downward Bias in Estimates of Liability-Scale Heritability in Genome-
wide Association Study Meta-analysis: A Simple Solution. Biol. Psychiatry 93, 29–36
(2023).
138. Meier, S. M. et al. Genetic Variants Associated With Anxiety and Stress-Related
Disorders: A Genome-Wide Association Study and Mouse-Model Study. JAMA
Psychiatry (2019) doi:10.1001/jamapsychiatry.2019.1119.
139. Derks, E. M., Thorp, J. G. & Gerring, Z. F. Ten challenges for clinical translation in
psychiatric genetics. Nat. Genet. 54, 1457–1465 (2022).
140. Flint, J. The genetic basis of major depressive disorder. Mol. Psychiatry (2023)
doi:10.1038/s41380-023-01957-9.
141. Mitchell, B. L. et al. Polygenic Risk Scores Derived From Varying Definitions of
Depression and Risk of Depression. JAMA Psychiatry 78, 1152–1160 (2021).
142. Cai, N. et al. Minimal phenotyping yields genome-wide association signals of low
specificity for major depression. Nat. Genet. 52, 437–447 (2020).
143. Nguyen, T.-D. et al. Genetic heterogeneity and subtypes of major depression. Mol.
Psychiatry 27, 1667–1675 (2022).
144. Mitchell, B. L. et al. The Australian Genetics of Depression Study: New Risk Loci
and Dissecting Heterogeneity Between Subtypes. Biol. Psychiatry (2021)
doi:10.1016/j.biopsych.2021.10.021.
145. Clements, C. C. et al. Genome-wide association study of patients with a severe
major depressive episode treated with electroconvulsive therapy. Mol. Psychiatry
26, 2429–2439 (2021).
146. Jermy, B. S., Glanville, K. P., Coleman, J. R. I., Lewis, C. M. & Vassos, E. Exploring
the genetic heterogeneity in major depression across diagnostic criteria. Mol.
Psychiatry 26, 7337–7345 (2021).
147. Howard, D. M. et al. Genetic stratification of depression in UK Biobank. Transl.
Psychiatry 10, 163 (2020).
148. Coleman, J. R. I. et al. The Genetics of the Mood Disorder Spectrum: Genome-wide
Association Analyses of More Than 185,000 Cases and 439,000 Controls. Biol.
Psychiatry (2019) doi:10.1016/j.biopsych.2019.10.015.
149. Cai, N., Choi, K. W. & Fried, E. I. Reviewing the genetics of heterogeneity in
depression: operationalizations, manifestations and etiologies. Hum. Mol. Genet. 29,
R10–R18 (2020).
150. Morneau-Vaillancourt, G. et al. The genetic and environmental hierarchical structure
of anxiety and depression in the UK Biobank. Depress. Anxiety 37, 512–520 (2020).
151. Forstner, A. J. et al. Genome-wide association study of panic disorder reveals
genetic overlap with neuroticism and depression. Mol. Psychiatry 26, 4179–4190
(2019).
152. Erhardt, A. et al. TMEM132D, a new candidate for anxiety phenotypes: evidence
from human and mouse studies. Mol. Psychiatry 16, 647–663 (2011).
153. Deckert, J. et al. GLRB allelic variation associated with agoraphobic cognitions,
increased startle response and fear network activation: a potential neurogenetic
pathway to panic disorder. Mol. Psychiatry 22, 1431–1439 (2017).
154. Cheesman, R., Rayner, C. & Eley, T. The Genetic Basis of Child and Adolescent

Page 48
47
Anxiety. Pediatric Anxiety Disorders 17–46 Preprint at https://doi.org/10.1016/b978-
0-12-813004-9.00003-7 (2019).
155. van Dongen, J., Slagboom, P. E., Draisma, H. H. M., Martin, N. G. & Boomsma, D. I.
The continuing value of twin studies in the omics era. Nat. Rev. Genet. 13, 640–653
(2012).
156. McLaughlin, K. A., Colich, N. L., Rodman, A. M. & Weissman, D. G. Mechanisms
linking childhood trauma exposure and psychopathology: a transdiagnostic model of
risk and resilience. BMC Med. 18, 96 (2020).
157. Waszczuk, M. A., Kotov, R., Ruggero, C., Gamez, W. & Watson, D. Hierarchical
structure of emotional disorders: From individual symptoms to the spectrum. J.
Abnorm. Psychol. 126, 613–634 (2017).
158. Lewis, S. J. et al. The epidemiology of trauma and post-traumatic stress disorder in
a representative cohort of young people in England and Wales. Lancet Psychiatry 6,
247–256 (2019).
159. McLaughlin, K. A. et al. Childhood adversities and first onset of psychiatric disorders
in a national sample of US adolescents. Arch. Gen. Psychiatry 69, 1151–1160
(2012).
160. Baldwin, J. R. et al. Childhood Maltreatment and Mental Health Problems: A
Systematic Review and Meta-Analysis of Quasi-Experimental Studies. Am. J.
Psychiatry appiajp20220174 (2023).
161. Koss, K. J. & Gunnar, M. R. Annual Research Review: Early adversity, the
hypothalamic–pituitary–adrenocortical axis, and child psychopathology. Journal of
Child Psychology and Psychiatry vol. 59 327–346 Preprint at
https://doi.org/10.1111/jcpp.12784 (2018).
162. Danese, A. & Baldwin, J. R. Hidden Wounds? Inflammatory Links Between
Childhood Trauma and Psychopathology. Annu. Rev. Psychol. 68, 517–544 (2017).
163. Kendler, K. S. & Baker, J. H. Genetic influences on measures of the environment: a
systematic review. Psychol. Med. 37, 615–626 (2007).
164. Wootton, R. E., Davis, O. S. P., Mottershaw, A. L., Wang, R. A. H. & Haworth, C. M.
A. Genetic and environmental correlations between subjective wellbeing and
experience of life events in adolescence. Eur. Child Adolesc. Psychiatry 26, 1119–
1127 (2017).
165. Peel, A. J. et al. A multivariate genetic analysis of anxiety sensitivity, environmental
sensitivity and reported life events in adolescents. J. Child Psychol. Psychiatry 64,
289–298 (2023).
166. Stein, M. B., Jang, K. L., Taylor, S., Vernon, P. A. & Livesley, W. J. Genetic and
environmental influences on trauma exposure and posttraumatic stress disorder
symptoms: a twin study. Am. J. Psychiatry 159, 1675–1681 (2002).
167. Pezzoli, P., Antfolk, J., Hatoum, A. S. & Santtila, P. Genetic Vulnerability to
Experiencing Child Maltreatment. Front. Genet. 10, 852 (2019).
168. Power, R. A. et al. Estimating the heritability of reporting stressful life events
captured by common genetic variants. Psychol. Med. 43, 1965–1971 (2013).
169. Dalvie, S. et al. Genomic influences on self-reported childhood maltreatment. Transl.
Psychiatry 10, 38 (2020).
170. Saudino, K. J., Pedersen, N. L., Lichtenstein, P., McClearn, G. E. & Plomin, R. Can
personality explain genetic influences on life events? J. Pers. Soc. Psychol. 72,
196–206 (1997).
171. Kendler, K. S., Karkowski, L. M. & Prescott, C. A. The assessment of dependence in

Page 49
48
the study of stressful life events: validation using a twin design. Psychol. Med. 29,
1455–1460 (1999).
172. Knafo, A. & Jaffee, S. R. Gene–environment correlation in developmental
psychopathology. Dev. Psychopathol. 25, 1–6 (2013).
173. Jaffee, S. R. & Price, T. S. Gene–environment correlations: a review of the evidence
and implications for prevention of mental illness. Mol. Psychiatry 12, 432–442
(2007).
174. Krapohl, E. et al. The high heritability of educational achievement reflects many
genetically influenced traits, not just intelligence. Proc. Natl. Acad. Sci. U. S. A. 111,
15273–15278 (2014).
175. McAdams, T. A., Gregory, A. M. & Eley, T. C. Genes of experience: explaining the
heritability of putative environmental variables through their association with
behavioural and emotional traits. Behav. Genet. 43, 314–328 (2013).
176. Barkhuizen, W., Dudbridge, F. & Ronald, A. Genetic overlap and causal
associations between smoking behaviours and mental health. Sci. Rep. 11, 14871
(2021).
177. Werme, J., van der Sluis, S., Posthuma, D. & de Leeuw, C. A. Genome-wide gene-
environment interactions in neuroticism: an exploratory study across 25
environments. Transl. Psychiatry 11, 180 (2021).
178. Turkheimer, E. Three Laws of Behavior Genetics and What They Mean. Curr. Dir.
Psychol. Sci. 9, 160–164 (2000).
179. Assary, E., Vincent, J. P., Keers, R. & Pluess, M. Gene-environment interaction and
psychiatric disorders: Review and future directions. Semin. Cell Dev. Biol. 77, 133–
143 (2018).
180. Peel, A. J. et al. Genetic and early environmental predictors of adulthood self-
reports of trauma. Br. J. Psychiatry 1–8 (2022).
181. Kendler, K. S. et al. Stressful life events, genetic liability, and onset of an episode of
major depression in women. Am. J. Psychiatry 152, 833–842 (1995).
182. Dick, D. M. Gene-environment interaction in psychological traits and disorders.
Annu. Rev. Clin. Psychol. 7, 383–409 (2011).
183. Pingault, J.-B. et al. Research Review: How to interpret associations between
polygenic scores, environmental risks, and phenotypes. J. Child Psychol. Psychiatry
63, 1125–1139 (2022).
184. Danese, A. & Widom, C. S. Objective and subjective experiences of child
maltreatment and their relationships with psychopathology. Nat Hum Behav 4, 811–
818 (2020).
185. Uliaszek, A. A. et al. The role of neuroticism and extraversion in the stress-anxiety
and stress-depression relationships. Anxiety Stress Coping 23, 363–381 (2010).
186. Assary, E., Zavos, H. M. S., Krapohl, E., Keers, R. & Pluess, M. Genetic architecture
of Environmental Sensitivity reflects multiple heritable components: a twin study with
adolescents. Mol. Psychiatry 26, 4896–4904 (2021).
187. Reuben, A. et al. Lest we forget: comparing retrospective and prospective
assessments of adverse childhood experiences in the prediction of adult health. J.
Child Psychol. Psychiatry 57, 1103–1112 (2016).
188. Gehrt, T. B., Berntsen, D., Hoyle, R. H. & Rubin, D. C. Psychological and clinical
correlates of the Centrality of Event Scale: A systematic review. Clin. Psychol. Rev.
65, 57–80 (2018).
189. Ogle, C. M., Siegler, I. C., Beckham, J. C. & Rubin, D. C. Neuroticism Increases

Page 50
49
PTSD Symptom Severity by Amplifying the Emotionality, Rehearsal, and Centrality
of Trauma Memories. J. Pers. 85, 702–715 (2017).
190. Rietschel, L. et al. Perceived stress has genetic influences distinct from neuroticism
and depression. Behav. Genet. 44, 639–645 (2014).
191. Belsky, J. & Pluess, M. Beyond diathesis stress: differential susceptibility to
environmental influences. Psychol. Bull. 135, 885–908 (2009).
192. Jolicoeur-Martineau, A. et al. Distinguishing differential susceptibility, diathesis-
stress, and vantage sensitivity: Beyond the single gene and environment model.
Dev. Psychopathol. 32, 73–83 (2020).
193. Cuijpers, P., Cristea, I. A., Karyotaki, E., Reijnders, M. & Huibers, M. J. H. How
effective are cognitive behavior therapies for major depression and anxiety
disorders? A meta-analytic update of the evidence. World Psychiatry vol. 15 245–
258 Preprint at https://doi.org/10.1002/wps.20346 (2016).
194. Rush, A. J. et al. Acute and longer-term outcomes in depressed outpatients
requiring one or several treatment steps: a STAR*D report. Am. J. Psychiatry 163,
1905–1917 (2006).
195. Bousman, C. A., Arandjelovic, K., Mancuso, S. G., Eyre, H. A. & Dunlop, B. W.
Pharmacogenetic tests and depressive symptom remission: a meta-analysis of
randomized controlled trials. Pharmacogenomics 20, 37–47 (2019).
196. Tansey, K. E. et al. Contribution of common genetic variants to antidepressant
response. Biol. Psychiatry 73, 679–682 (2013).
197. Pain, O. et al. Identifying the Common Genetic Basis of Antidepressant Response.
Biol Psychiatry Glob Open Sci 2, 115–126 (2022).
198. David, D., Cristea, I. & Hofmann, S. G. Why Cognitive Behavioral Therapy Is the
Current Gold Standard of Psychotherapy. Front. Psychiatry 9, 4 (2018).
199. Powers, M. B., de Kleine, R. A. & Smits, J. A. J. Core Mechanisms of Cognitive
Behavioral Therapy for Anxiety and Depression: A Review. Psychiatr. Clin. North
Am. 40, 611–623 (2017).
200. Newby, J. M., McKinnon, A., Kuyken, W., Gilbody, S. & Dalgleish, T. Systematic
review and meta-analysis of transdiagnostic psychological treatments for anxiety
and depressive disorders in adulthood. Clin. Psychol. Rev. 40, 91–110 (2015).
201. Garc�a-Escalera, J., Chorot, P., Valiente, R. M., Reales, J. M. & Sand�n, B. Efficacy
of transdiagnostic cognitive-behavioral therapy for anxiety and depression in adults,
children and adolescents: A meta-analysis. Revista de Psicopatolog�a y Psicolog�a
Cl�nica 21, 147–175 (2016).
202. Tanguay-Sela, M. et al. A systematic meta-review of patient-level predictors of
psychological therapy outcome in major depressive disorder. J. Affect. Disord. 317,
307–318 (2022).
203. Purves, K. L. et al. Evidence for distinct genetic and environmental influences on
fear acquisition and extinction. Psychol. Med. 1–9 (2021).
204. Hettema, J. M., Annas, P., Neale, M. C., Kendler, K. S. & Fredrikson, M. A twin
study of the genetics of fear conditioning. Arch. Gen. Psychiatry 60, 702–708
(2003).
205. Zanardi, R., Carminati, M., Attanasio, F., Fabbri, C. & Serretti, A. Genetics of
nonpharmacological treatments of depression. Psychiatr. Genet. 33, 1–7 (2023).
206. Lester, K. J. & Eley, T. C. Therapygenetics: Using genetic markers to predict
response to psychological treatment for mood and anxiety disorders. Biol. Mood
Anxiety Disord. 3, 4 (2013).

Page 51
50
207. Keers, R. et al. A Genome-Wide Test of the Differential Susceptibility Hypothesis
Reveals a Genetic Predictor of Differential Response to Psychological Treatments
for Child Anxiety Disorders. Psychother. Psychosom. 85, 146–158 (2016).
208. Lester, K. J. et al. Non-replication of the association between 5HTTLPR and
response to psychological therapy for child anxiety disorders. Br. J. Psychiatry 208,
182–188 (2016).
209. Rayner, C. et al. A genome-wide association meta-analysis of prognostic outcomes
following cognitive behavioural therapy in individuals with anxiety and depressive
disorders. Transl. Psychiatry 9, 150 (2019).

Page 52
51
Chapter 2. Exploring the genome-wide genetic
overlap between anxiety and fear disorders
This chapter is a manuscript that is in preparation for peer-review. Supplementary materials
for this chapter, as detailed in the text, are included in Appendix A.
Authors: Abigail R. ter Kuile1,2, Brittany L. Mitchell3, Genevi�ve Morneau-Vaillancourt1, Sang
Hyuck Lee1, Helena L. Davies1, Jessica Mundy1,2, Alicia J. Peel1, Megan Skelton1,2,
Christopher H�bel1,2,9, Molly R. Davies1,2, Jonathan R. I. Coleman1,2, Anna E. F�rtjes1, Zain
Ahmad1, Yuhao Lin1, Brett N. Adey1,2, Thomas McGregor1, Kirstin Purves1,2, Alish Palmos1,
Johan Zvrskovec1,2, Matthew Hotopf1,2,8, Gursharan Kalsi1,2, Ian R. Jones10, Daniel J. Smith11,
David Veale1,2,8, James T. R. Walters10, Ch�rie Armour5, Colette R. Hirsch1,2,8, Andrew M.
McIntosh11, Naomi R. Wray12,13, Sarah E. Medland3, Enda M. Byrne6, Nicholas G. Martin3,
Gerome Breen1,2, Thalia C. Eley1,2
1. Institute of Psychiatry, Psychology and Neuroscience, King's College London,
Denmark Hill, Camberwell, London, UK
2. National Institute for Health and Care Research (NIHR) Maudsley Biomedical
Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
3. QIMR Berghofer Medical Research Institute, Brisbane, Australia
4. School of Biomedical Sciences, Faculty of Medicine, The University of Queensland,
Brisbane, Australia.
5. Research Centre for Stress, Trauma, and Related Conditions (STARC), School of
Psychology, Queen’s University Belfast (QUB), Belfast, Northern Ireland, UK
6. Child Health Research Centre, The University of Queensland, Brisbane, Australia
7. Brain and Mind Centre, The University of Sydney, Sydney, New South Wales,
Australia
8. South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks
Orchard Road, Beckenham, Kent, UK
9. National Centre for Register-based Research, Aarhus Business and Social Sciences,
Aarhus University, Aarhus, Denmark
10. MRC Centre for Neuropsychiatric Genetics and Genomics, National Centre for
Mental Health, Cardiff University, Cardiff,UK
11. Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh,
Edinburgh, UK
12. Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
13. Queensland Brain Institute, The University of Queensland, Brisbane, Australia

Page 53
Abstract
Background: Twin studies have consistently shown a high genetic overlap amongst anxiety
disorders and depression that contributes to the internalising spectrum. Research has also
identified modest genetic specificity to fear-based anxiety disorders, not shared with general
anxiety and depression (often grouped as distress disorders). Due to the lack of datasets with
detailed phenotyping of all five anxiety disorders, genome-wide analysis of anxiety has
typically been limited to “any anxiety diagnosis”. Further genome-wide evidence is needed to
establish if fear-based disorders are genetically distinct from distress disorders.
Methods: We undertook a genome-wide association study (GWAS) meta-analysis of fear-
based disorders (panic, social anxiety disorder, specific phobia, and agoraphobia) and
generalised anxiety disorder (GAD). Cases and controls were defined using a combination of
brief single-item and detailed symptom-based diagnoses from three large datasets. We
explored two approaches to define controls. First, we screened for any anxiety disorder and
depression. Second, we screened specifically for fear or GAD. We estimated genetic
correlations between fear and GAD and compared their correlations with broad domains of
other complex traits.
Results: Our GWAS meta-analyses identified a total of three independent loci associated with
fear (up to 30,861 Ncases; 157,951 Ncontrols) and four with GAD (up to 54,928 Ncases; 117,320
Ncontrols). The genetic correlation between fear and GAD was not significantly different from
one, except for when excluding a depression-enriched dataset and screening controls
specifically (rg = 0.85; P = 4.57 � 10-3). Most complex traits did not have a significantly different
genetic correlation with fear versus GAD, including depression. The exceptions to this
included general cognitive ability, educational attainment, and coronary artery disease with
stronger negative genetic correlations with fear than GAD. Bipolar disorder type I, anorexia
nervosa, and neuroticism had stronger positive genetic correlations with GAD than fear.
Conclusions: Our findings partially support a distress-fear genetic distinction. However, we
found stronger evidence for an overarching genetic liability to internalising psychopathology
that drives comorbidity across anxiety disorders and depression.

Page 54
53
Introduction
Anxiety disorders, including panic disorder, agoraphobia, specific phobia, social anxiety
disorder, and generalised anxiety disorder (GAD), are among the most common mental health
conditions, with an international lifetime prevalence of 16%1,2. The core symptoms of anxiety
disorders are persistent and excessive fear and worry, accompanied by impairing features
such as avoidance behaviours, physiological arousal, and panic attacks3. Individual disorder
diagnoses across the anxiety spectrum are based upon their distinct clinical features, although
high rates of comorbidity exist between them4,5. GAD is characterised by excessive and
chronic worry about a broad range of future potential threats. Panic disorder is an extreme
version of a flight or fight response with repeated experiences of a sudden unexpected feeling
of fear in the absence of a threat (i.e. panic attack). Recurrent, unexpected panic attacks are
necessary for a diagnosis of panic disorder. Panic attacks, however, are also transdiagnostic
and can occur in certain situations, as in phobic disorders. The phobias, including
agoraphobia, social anxiety disorder and specific phobia, are characterised by excessive fear
and avoidance of particular signals of perceived threat4,6. As they are all characterised by fear,
panic disorder and the phobias are often grouped together as ‘fear-based’ disorders.
Shared risk factors for anxiety disorders include being female, exposure to stressful
experiences, and a family history of anxiety and depression3. Familial aggregation of these
disorders reflects a partial genetic basis, with twin study estimates of heritability ranging from
20-60%3,7,8. Twin studies consistently show that most genetic influences on anxiety disorders
overlap, contributing to a broad anxiety-related genetic factor, which has considerable overlap
with genetic risk for depression9–12. However, evidence suggests this higher-order internalising
genetic liability can be differentiated into lower-order genetic components. Fear-based
disorders are reported to be partly genetically distinct from those characterised by distress
(GAD and depression), forming two distinct but correlated genetic liability fear-distress
subfactors9,13. Overall, twin studies show that anxiety disorders mostly share common genetic
factors but also have some genetic specificity.
Although twin studies have been crucial in revealing the overall genetic structure of the anxiety
disorder spectrum, they do not identify the specific genetic variants that contribute to the
heritability and high genetic overlap between the disorders. Genome-wide association studies
(GWAS) have begun to identify common genetic variants underpinning anxiety disorders.
Fewer GWAS have been performed in anxiety disorders than in other psychiatric disorders14.
However, recent developments have enabled several large-scale GWAS of broadly defined
anxiety and GAD symptoms15–18. Such studies have identified significant genetic correlations
with all other psychiatric disorders, including depression, schizophrenia, bipolar disorder, and

Page 55
54
attention deficit hyperactivity disorder (ADHD)15–17. Consistent with evidence from twin
studies9, preliminary genome-wide findings indicate that a lifetime diagnosis of any fear-based
disorder (as a grouping) and depression are less genetically correlated with one another than
they each are with lifetime GAD19. However, the common genetic variants associated with
fear-based disorders remain unclear, which limits our understanding of their genetic overlap
at the molecular level.
Previous GWAS of fear-based disorders have been underpowered for loci discovery and to
assess genetic correlations with a broad range of traits19–21. Such analyses would provide
more molecular genetic evidence and fine-grained detail to the current hierarchical structures
of the psychiatric spectrum, which could have implications for improving diagnostic systems22.
A formal comparison of fear and GAD genetic correlations with other psychiatric, behavioural,
cognitive, or health traits could suggest transdiagnostic or subtype-specific mechanisms.
Further well-powered genome-wide evidence of the genetic distinction between distress and
fear is needed in the broader context of their relationships with other complex traits.
The way in which anxiety disorders are measured is an important consideration when
identifying genetic similarities and differences. Dealing with comorbidity is a particular
challenge in identifying anxiety-specific genetic influences, as anxiety disorders are among
the most highly comorbid disorders across psychopathology23. Screening controls for co-
occurring traits in GWAS could inflate genetic correlations24. A comparison of different control
screening approaches is needed. Furthermore, genetic influences captured by using brief
phenotyping measures may be less specific to the focal trait than those captured via detailed
phenotyping, reflecting more general psychopathology25,26. The previous GWAS reporting
distress-fear genetic correlations was limited to brief, single-item reports of receipt of a
diagnosis by a health professional for each fear disorder19. A recent study found that rates of
fear-based disorder diagnoses were lower when using brief self-report diagnoses than
detailed symptom-based measures, and observed the opposite pattern for GAD27. Since
identifying genetic variants requires large sample sizes, a trade-off exists between maximising
sample size and the level of detail in phenotyping. One way to achieve larger sample sizes
while retaining phenotypic specificity is by combining detailed symptom-based diagnoses with
brief single-item self-report diagnoses28.
Here, we aimed to assess the shared and non-shared genetic influences on fear-based
disorders and GAD at the level of common genetic variants. We increased sample size and
thus power for loci-discovery and genetic correlation analyses by combining detailed
symptom-based diagnoses with brief single-item diagnoses. We examined differences in
genetic correlations between fear-based disorders and GAD with a range of other complex

Page 56
55
traits, and also assessed broad, and more specific approaches to screening controls. Our
study represents progress in the anxiety disorder genomics field and adds further detail on the
distress-fear distinction at the genome-wide level.

Page 57
56
Methods
Phenotypes
Anxiety disorder phenotypes
Analyses were part of a pre-registered plan on the Open Science Framework (accessible at
https://tinyurl.com/OSFfearGAD). We conducted GWAS meta-analyses of two lifetime anxiety
disorder phenotypes: i) fear-based disorders (referred to as ‘fear’ hereafter; panic,
agoraphobia, specific phobia and social anxiety disorder; N cases = 30,861) and ii) GAD (N
cases = 54,928). We used the term “panic” instead of “panic disorder” as brief diagnostic
measures of self-reported panic attacks were used in addition to brief and detailed diagnostic
measures of panic disorder. This increased power and data harmonisation, as the UK Biobank
Mental Health Questionaire only had brief measures on panic attacks29 (Supplementary
Tables 1 & 2). Although panic attacks are not specific to panic disorder, the brief diagnostic
measure of panic attacks has shown reasonable agreement with detailed diagnostic measures
of panic disorder27. Five samples were included in the GWAS meta-analyses. The five
samples resulted in three case and control datasets, as detailed below (see Table 1 for a
summary of sample sizes). Individuals were screened for an anxiety disorder using a
combination of detailed symptom-based diagnoses and brief, self-report diagnoses. Fear
cases were defined as participants who endorsed at least one fear disorder.
Detailed diagnostic measures
Detailed symptom-based measures of anxiety disorders were derived using previously
described algorithms27. Cases for each phenotype were defined as those who met DSM-5
criteria for a lifetime symptom-based disorder diagnosis. Each anxiety disorder was assessed
using an online, self-report questionnaire version of the Composite International Diagnostic
Interview short-form (CIDI-SF)30,31. The availability of questionnaires in each study is
summarised in Supplementary Table 1.
Brief diagnostic measures
Individuals meeting case status on brief measures endorsed a single-item self-report question
for the corresponding disorder, e.g. “Have you ever been diagnosed with one or more of the
following mental health problems by a professional, even if you do not have it currently?”. The
exact phrasing of the question and responses in each study, as well as mapping to disorders,
are provided in Supplementary Table 2.

Page 58
57
Control groups
We explored two different ways of defining control groups. First, controls were screened for
any anxiety disorder or depression (fearanx-dep and GADanx-dep). Due to the high genetic
correlation between these disorders19, this was the most powerful approach for loci discovery.
Detailed and brief measures were used to screen controls for any anxiety disorder
(Supplementary Tables 1 & 2). Controls were screened for depression using brief, self-report
diagnoses in all datasets. Second, to better distinguish genetic differences between GAD and
fear, we defined specifically screened controls whereby participants were only screened for
the specific disorder being analysed (fearspecific and GADspecific). This is because both fear and
GAD have high genetic overlap with one another and with depression, thus screening controls
for any anxiety disorder and depression could further increase the genetic correlation between
them. Fearspecific GWAS controls were only screened for any fear disorder, and GADspecific
GWAS controls were only screened for GAD where possible. Screening controls specifically
for only GAD or only fear-based disorders was not possible in the QIMR dataset as we were
limited to using a broad, single-item self-report diagnosis of “anxiety”. Controls were not
screened for the presence of other psychiatric disorders as this can bias genetic correlation
estimates24.
Samples
The GLAD Study and COPING Study (GLAD+ dataset)
Data from two studies within the National Institute for Health and Care Research (NIHR)
BioResource were included. Cases were primarily ascertained from the Genetic Links to
Anxiety and Depression (GLAD) study32. The GLAD study is an online research platform
launched in September 2018 to recruit participants with lifetime anxiety and/or depressive
disorder. Recruitment is ongoing and is open to individuals residing in the UK and aged at
least 16 years. Initial recruitment involved a widespread media campaign, followed by ongoing
social media advertising and participating NHS trusts and GP practices. Controls for analyses
with GLAD cases were ascertained from a longitudinal survey run by the GLAD team,
collecting data within GLAD and other NIHR BioResource cohorts called the COVID-19
Psychiatric and Neurological Genetics (COPING) study33. The phenotyping in COPING for
anxiety disorders was identical to GLAD. A proportion of COPING study participants met the
criteria for an anxiety disorder and were defined as cases (Table 1). Cases and controls in the
GLAD and COPING studies were defined using both brief and detailed measures of all five
anxiety disorders. We refer here to the GLAD and COPING combined study samples as the
GLAD+ dataset.

Page 59
58
Queensland Institute of Medical Research (QIMR) Berghofer dataset
A second unrelated set of case-control analyses included data from two Australian QIMR
Berghofer studies. Cases were obtained from the Australian Genetics of Depression Study
(AGDS), launched in September 2016, to collect genetic and phenotypic data on individuals
with a lifetime experience of depression31. Recruitment is ongoing and has been conducted
via pharmaceutical prescription history provided by the Australian government and through
media campaigns. Participation is open to individuals aged at least 18 years across Australia.
AGDS cases were defined using detailed and brief self-report measures of all five anxiety
disorders. Controls for analyses with AGDS cases were obtained from the QIMR Berghofer
QSkin Sun & Health Study34, defined using a brief self-report measure of any anxiety disorder.
The UK Biobank dataset
The UK Biobank is a population-based dataset of over 500,000 participants aged between 40
and 69 recruited from 2006 to 201035. In 2017, a subset of participants (N = 157,366)
completed the online follow-up Mental Health Questionnaire (MHQ) assessing mental health
and well-being through self-report measures29. Participants were defined as cases or controls
using brief measures of fear disorders and brief and detailed measures of GAD.

Page 60
59
Table 1. Sample sizes in each study, GWAS dataset, and meta-analysis of fear-based disorders and GAD.
Control criteria
N dataset total
(Ncases + Ncontrols)
Anxiety disorder
GWAS
Datasets
Study
Sample
Ncases
Anx-dep controls
Specific controls
Anx-
dep
Specific
Ncontrols
Neffective
Ncontrols
Neffective
Fear
GLAD+
GLAD
12,243
-
13,300
-
16,961
17,848
19,626
COPING
1,186
4,419
6197
UKB
10,789
83,236
38,204
137,209
40,010
94,025
147,998
QIMR
AGDS
6,643
-
17,457
-
18,241
19,365
21,188
Qskin
-
12,722
14,545
Total meta-analysis N
(GLAD+, UKB & QIMR)
30,861
100,377
68,960
157,951
75,212
131,238
188,812
Total meta-analysis N
(GLAD+ & UKB)
24,218
87,655
51,504
143,406
56,971
111,873
167,624
GAD
GLAD+
GLAD
14,339
-
13,857
-
19,606
20,452
23,093
COPING
1,694
4,419
7060
UKB
26,067
83,236
79,402
95,715
81,950
109,303
121,782
QIMR
AGDS
12,828
-
25,550
-
27,265
25,550
27,373
Qskin
-
12,722
14,545
Total meta-analysis N
GLAD+, UKB & QIMR)
54,928
100,377
118,808
117,320
128,822
155,305
172,248
Total meta-analysis N
(GLAD+ & UKB)
42,100
87,655
93,259
102,775
101,556
129,755
144,875
Numbers shown in bold are total sample size used in GWAS meta-analyses. Effective sample size was calculated as: Neffective = 4/[(1/ncase) +
(1/ncontrol)], which converts the power of a study sample size to one with a balanced case-control ratio (i.e. equivalent to a study with a sample
prevalence of 50%, with Ncases = Neffective and Ncontrols = Neffective).

Page 61
60
Genome-wide association analyses
Genotyping and quality control was performed separately in each study (Supplementary
Materials). Common genetic variants in the GLAD+ and QIMR datasets were imputed to the
Top Med imputation panel (Version R2 on GRC38) and the UK Biobank to the Haplotype
Reference Consortium and the UK10K Consortium reference panels. GWAS analyses were
restricted to common genetic variants (MAF > 1%) and high confidence-imputed variants
(INFO score > 0.3 for TOPMed imputed variants; > 0.4 for UK Biobank).
We ran four anxiety disorder GWASs; fearspecific, GADspecific, fearanx-dep and GADanx-dep. A
separate GWAS was conducted for each of the three datasets (GLAD+, UK Biobank, QIMR).
Sample sizes for each GWAS are shown in Table 1. The software REGENIE v3.1.3 was used
for analyses in the GLAD+ and the UK Biobank datasets, whilst SAIGE v0.44 was used in the
QIMR dataset36,37. The first ten principal components and genotyping batch were included as
covariates. Additionally, the assessment centre was included as a covariate for GWAS in the
UK Biobank. GWAS were limited to participants of European-associated genetic ancestry
clusters, defined using principal component analysis (PCA).
Meta-analyses and annotation
We used the software METAL to perform inverse-variance weighted meta-analyses of each
anxiety disorder phenotype38. In our primary analyses, we meta-analysed GWAS results from
the GLAD+, QIMR Berghofer, and UK Biobank datasets (termed full meta-analysis
throughout). As all cases from the AGDS also had comorbid depression, we conducted a
second set of meta-analyses excluding the QIMR datasets (referred to as GLAD+ and UKB
meta-analysis). A depression-enriched dataset might increase depression/distress genetics in
anxiety disorder GWAS, elevating the genetic correlation between fear, GAD, and depression.
Meta-analysis sample sizes are shown in Table 1. We restricted common genetic variants to
those overlapping across datasets in each meta-analysis. Associated variants were mapped
and annotated using FUMA v1.4.0 (with default parameters applied) and the UKB release2b
10K European reference panel39. MAGMA v1.08 was used with default parameters to identify
gene-level and biological pathway associations. Gene-level association analysis aggregates
the combined effect of associated common genetic variants at the level of the gene. Results
from gene-level analyses were then applied to gene-set analysis to conduct biological pathway
analyses40.

Page 62
61
SNP-based heritability
We estimated the heritability captured by common genetic variants (h2
SNP) of fear and GAD.
Population prevalences for liability scale estimates were based on the assumption of accurate
sampling from the COPING study, in which both detailed and brief measures were available
to define cases and controls for all five anxiety disorders (Supplementary Table 3). GWAS
meta-analysis summary results were used in LDSC regression to estimate h2
SNP
41.
Genetic correlations
Using LDSC regression42, we calculated genetic correlations (rg) between i) fear and GAD and
ii) fear and GAD with 345 external complex traits respectively. Traits were considered
sufficiently powered for genetic correlation analysis if they had a GWAS mean Χ2 > 1.02 and
a heritability Z score > 4, calculated in LDSC regression. We tested if the genetic correlation
between fear and GAD was significantly different from 0 (using default parameters in bivariate
LDSC regression) and significantly different from 1 (calculated in R using the chi-squared
distribution function and [(|rg|−1)/se]2 ). To correct for multiple testing, we applied a Bonferroni
corrected P-value threshold (𝛼[0.05] / the number of fear-GAD rg tested [4] = P  ≤ 0.0125).
Fear and GAD GWAS with a genetic correlationsignificantly different from 1 were then tested
for genetic correlations with external traits. We selected external traits in a hypothesis-free
manner and applied a Bonferroni-corrected significance threshold (0.05 / number of external
traits tested [345] = P ≤ 1.45 � 10-4). We tested for differences between fear versus GAD in
terms of their respective genetic correlations with external traits using the block-jackknife
method implemented in LDSC regression42,43. Fear and GAD were considered to have a
significantly different genetic correlation with an external trait if the P-value derived from the
block-jackknife Z statistic result exceeded the Bonferroni-corrected threshold
(P  ≤ 1.45 � 10-4).

Page 63
62
Results
Genome-wide association meta-analyses
Manhattan plots for the most well-powered GWAS are shown in Figure 1 (fearanx-dep and
GADanx-dep in the full meta-analysis). Manhattan plots for all other GWAS with genome-wide
significant loci are shown in Supplementary Figures 2, 4, 14 and 16. We found little evidence
of confounding in all GWAS (LDSC intercept = 0.96-1.04). LDSC intercepts >1 indicate some
inflation, which is expected as the GWAS sample size increases44. The majority of inflation in
our meta-analyses was due to polygenicity (90-97%), as indicated by LDSC attenuation ratio
calculations. The genomic inflation factor, LDSC intercept, attenuation ratio, and Q-Q plots for
each GWAS are shown in Supplementary Figures 1, 3, 12, 13 and 15. Regional plots for
independent genome-wide significant loci are also in Supplementary Figures 5-11.
Fear-based disorders
Across the four GWASs, we found a total of three independent genome-wide significant loci
associated with fear. In each fear GWAS, a different locus was identified, except for fearspecific
in the GLAD+ and UKB meta-analysis, where no loci reached P < 5 � 10−8. The locus with the
most significant lead variant was rs10047892 on chromosome 14 in the fearanx-dep GWAS in
the full meta-analysis (P = 2.99 � 10−8; Figure 1, upper panel). The fearspecific GWAS in the
full meta-analysis showed a locus on chromosome 5 as genome-wide significant (lead variant
rs3996354; P = 4.71 � 10−8). A locus on chromosome 1 with lead variant rs11576254 was
found in the fearanx-dep
GWAS in the GLAD+ and UKB meta-analysis
(P = 4.70 � 10−8; Table 2).
Generalised anxiety disorder
In all, we identified four independent genome-wide significant loci associated with GAD
(P < 5 � 10−8), although not all four loci reached genome-wide significance in each GAD
GWAS. The most significantly associated locus was on chromosome 9 (lead variant
rs10120318; P = 3.1 � 10−13; Figure 1, lower panel) and was genome-wide significant in all
four GAD GWASs (Table 2). The locus on chromosome 6 was the second most significant
(lead variant rs3858; P = 1.1 � 10−9; Figure 1, lower panel) and reached genome-wide
significance in all GWAS meta-analyses of GAD, except for GADspecific in the UKB and GLAD+
meta-analysis. The locus on chromosome 2 exceeded genome-wide significance in the
GADspecific GWAS in the full meta-analysis (lead variant rs11688767; P = 4.20 � 10−8), and a
locus on chromosome 5 in the GADspecific GWAS in the UKB and GLAD+ meta-analysis (lead
variant rs3095951; P = 4.33 � 10−8; Table 2).

Page 64
63
Figure 1: Genome-wide association study Manhattan plots for anxiety disorder
phenotypes meta-analysed across the GLAD+, QIMR and UKB datasets.
Fear-based disorder GWAS (upper panel) and GAD GWAS (lower panel) results from
phenotypes with controls screened for any anxiety disorder and depression. Dashed line
red; common genetic variant genome-wide significance threshold (P < 5 � 10−8), black;
suggestive significance threshold (P < 1 � 10−5).

Page 65
64
Table 2: Independent genome-wide significant loci associated with anxiety disorder phenotypes
Anxiety
disorder Datasets
Control
screening
criteria
Locus
no. Region CHR Lead SNP Base pair A1:A2 Func; gene (kb)
P
OR
N cand.
SNPs
Ind. Sig.
SNPs
Previous report in
GWAS catalog
Fear
QIMR,
GLAD+ &
UKB
Anx-dep
1 14q24.3 14 rs10047892 75111346 T:C
intergenic;
AREL1 (8793)
2.99E-08 1.07 26 rs10047892
EA, worry, systolic blood
pressure
Specific
2 5q31.3
5 rs3996354 143264699 A:G
intergenic; CTB-
57H20.1 (56361)
4.71E-08 0.93 20 rs3996354
Novel
GLAD+ &
UKB
Anx-dep
3 1p34.1
1 rs11576254 44835833 T:C
intergenic; ERI3
(14900)
4.70E-08 1.09 64 rs11576254
None at genome-wide
significance. At suggestive
threshold: metabolite
levels, reaction time,
anxiety
Specific
-
-
-
-
-
-
-
-
-
-
GAD
QIMR,
GLAD+ &
UKB
Anx-dep
4
9p23
9 rs10120318 11645069 A:T
intergenic; RP11-
23D5.1 (368754)
3.07E-13 0.93 301
rs10120318;
rs11515172
Neuroticism, depression,
wellbeing, any anxiety
disorder, GAD symptoms,
worry, BMI
5 6p22.1
6
rs385816 29480224 A:G
intergenic;
XXbac-
BPG13B8.10
(1890)
1.08E-09 0.92 184 rs385816
SCZ, HDL, BIP, cognitive
performance, depression,
cardiometabolic &
hematological traits, lung
cancer, worry, smoking,
neuroticism
Specific
4
9p23
9 rs10960024 11616820 C:G
intergenic; RP11-
23D5.1 (340505)
7.17E-13 0.93 301
rs10960024;
rs11515172
As locus 4 above
5 6p22.1
6
rs385816 29480224 A:G
intergenic;
XXbac-
BPG13B8.10
(1890)
7.32E-09 0.93 184 rs385816
As locus 5 above
6 2p16.1
2 rs11688767 57988194 A:T
ncRNA intronic;
CTD-2026C7.1
(0)
4.20E-08 0.95 19 rs11688767
Cross-disorder, sleep,
SCZ, cognitive ability,
depression, neuroticism,
epilepsy, neuroticism

Page 66
65
Table 2: Independent genome-wide significant loci associated with anxiety disorder phenotypes
Anxiety
disorder Datasets
Control
screening
criteria
Locus
no. Region CHR Lead SNP Base pair A1:A2 Func; gene (kb)
P
OR
N cand.
SNPs
Ind. Sig.
SNPs
Previous report in
GWAS catalog
GAD
GLAD+ &
UKB
Anx-dep
4
9p23
9 rs17189482 11513617 T:G
intergenic; RP11-
23D5.1 (237302)
1.24E-11 1.09 295
rs17189482;
rs11515172
As locus 4 above
5 6p22.1
6 rs3117427 29274136 T:C
upstream;
OR14J1 (266)
8.70E-09 1.09 219 rs3117427
As locus 5 above
7
5q34
5 rs3095951 164616460 A:G
intergenic; CTC-
340A15.2
(17810)
4.33E-08 1.06 142 rs3095951
Any anxiety disorder,
depression, neuroticism,
wellbeing, BMI
Specific
4
9p23
9 rs10960024 11616820 C:G
intergenic; RP11-
23D5.1 (340505)
1.63E-11 0.92 250 rs10960024
As locus 4 above
Locus no. = annotated locus number; CHR = chromosome; A1 = effect allele; A2 = noneffect allele; Func gene (kb) = functional consequence of the SNP on the nearest gene
with distance in kilobase; OR = odds ratio; N cand. SNPs = Number of genome-wide candidate SNPs that are in LD (r2 = 0.6) with Ind. Sig. SNPs; Ind. Sig. SNPs = independent
significant SNPs in the locus. Previous report = GWASs of other phenotypes that reported a genome-wide association with one or more of the candidate SNPs identified in this
study (full results shown in Supplementary Tables 4 & 5for fear and GAD loci, respectively). EA = educational attainment; BMI = body mass index; SCZ = schizophrenia, HDL
= high-density lipoprotein cholesterol, BIP = bipolar disorder.

Page 67
66
Gene-level and gene-set association analyses
Fear-based disorders
Three genes were significantly associated with fear-based disorders after Bonferroni
correction for the number of genes tested (see Table 3 for P-value thresholds). The gene
SNX29 on chromosome 16 was significantly associated with fear in all GWASs. The gene
YWHAG on chromosome 7 was only significantly associated with both fear GWAS in the full
meta-analysis, whilst the gene ERI3 on chromosome 1 was only significantly associated with
both fear GWAS in the GLAD+ and UKB meta-analysis (Table 3). The gene ERI3 includes
the SNP-level genome-wide significant locus 1p34.1. No gene sets were significantly
associated with fear (Bonferroni correction threshold for 10678 gene sets = P ≤ 4.68 � 10−6).
Generalised anxiety disorder
A total of three genes were significantly associated with GAD. All three genes were identified
in the GADanx-dep GWAS in the full meta-analysis. The most significantly associated gene was
TMEM106B on chromosome 7. This gene was also found in GADspecific GWAS in the full meta-
analysis and the GADanx-dep GWAS in the GLAD+ and UKB meta-analysis. The gene TRIM31
on chromosome 6 was also associated with GADspecific GWAS in the full meta-analysis. This
gene region includes common genetic variants on locus 6p22.1 identified in SNP-level
association analyses. The gene SORCS3 on chromosome 10 was only associated with
GADanx-dep GWAS in the full meta-analysis (Table 3). No gene sets were significantly
associated with GAD (P ≤ 4.68 � 10−6).

Page 68
67
Table 3: Gene-level associations with anxiety disorder phenotypes.
Anxiety
disorder Datasets
Control
screening
criteria
Gene
symbol CHR
N
SNPS Z
P
P Bonf.
Threshold
(0.05/
N genes) Previous report in GWAS catalog
Gene description
Fear
QIMR,
GLAD+ &
UKB
Anx-dep
SNX29
16 1560 5.09 1.77E-07
2.702E-6
(18502
genes)
EA, insomnia, smoking, IQ
Sorting nexin 29
YWHAG
7
46 4.67 1.49E-06
Blood protein levels. Suggestive
significance: SCZ, multiple sclerosis
Tyrosine 3-
monooxygenase/tryptophan 5-
monooxygenase activation protein
gamma
Specific
SNX29
16 1560 4.97 3.31E-07 2.702E-
6
(18502)
As above
As above
YWHAG
7
46 4.64 1.75E-06
As above
As above
GLAD+ &
UKB
Anx-dep
SNX29
16 1561 5.17 1.19E-07
2.698E-6
(18531)
As above
As above
ERI3
1 159 5.05 2.23E-07
EA, alcohol consumption
ERI1 exoribonuclease family member
3
Specific
SNX29
16 1562 5.07 1.96E-07 2.698E-6
(18531)
As above
As above
ERI3
1 159 4.62 1.94E-06
As above
As above
GAD
QIMR,
GLAD+ &
UKB
Anx-dep
TMEM106B 7 125 4.98 3.22E-07
2.702E-6
(18503)
Depression, EA, wellbeing, insomnia,
Alzheimer’s, neuroticism, GAD, PTSD,
HDL
Transmembrane protein 106B
SORCS3 10 1422 4.92 4.33E-07
EA, depression, blood pressure,
externalising behaviours, CAD,
insomnia, neuroticism, smoking, ADHD
Sortilin related VPS10 domain
containing receptor 3
TRIM31
6
45 4.81 7.74E-07
Cholesterol, depression, insomnia, IQ
Tripartite motif containing 31
Specific
TMEM106B 7 125 4.71 1.27E-06 2.702E-6
(18503)
As above
As above
TRIM31
6
45 4.58 2.31E-06
As above
GLAD+ &
UKB
Anx-dep TMEM106B 7 125 4.58 2.27E-06
2.698E-6
(18529)
As above
As above
Specific
-
-
-
-
-
2.698E-6
(18530)
-
-
Gene-level association analyses conducted in MAGMA. CHR = chromosome; Previous report = GWASs of other phenotypes that report genome-wide significant
loci that map onto the gene-level associations identified in this study (Supplementary Tables 6 & 7); EA = educational attainment; IQ = general cognitive ability;
SCZ = schizophrenia; GAD = generalised anxiety disorder; PTSD = post-traumatic stress disorder; HDL = high-density lipoprotein cholesterol; CAD = coronary
artery disease; ADHD = Attention deficit hyperactivity disorder.

Page 69
68
SNP-based heritability
We report results here for the most powerful GWAS in the full meta-analyses, as measured
by GWAS mean Χ2, and present results for other GWAS in the supplementary. We identified
modest liability-scale LDSC h2
SNP for Fearanx-dep of 9.6% (SE = 0.006, mean Χ2 = 1.17) and
10% for GADanx-dep (SE = 0.006, mean Χ2 = 1.25), assuming a population prevalence of 8.8%
and 12.9%, respectively. Population prevalence was based on COPING study sample
prevalence (Supplementary Table 3). We note that this is not a population-based study, and
a proportion of participants were recruited through an inflammatory bowel disease study45.
However, COPING study anxiety disorder prevalence estimates were within the range of
epidemiological studies, which report broad estimates for panic disorder (1-5%), agoraphobia
(1-3%), social anxiety disorder (3-13%), specific phobia (8-14%), and GAD (3-13%)46,47. To
our knowledge, no previous epidemiological studies have estimated the population prevalence
of fear-based disorders as a group. Supplementary Table 8 shows h2
SNP liability-scale
estimates calculated from prevalences 10% higher and lower than COPING study sample
prevalences. Supplementary Table 8 also reports h2
SNP liability-scale estimates for
phenotypes with specifically screened controls, and in the meta-analysis excluding QIMR.
Genetic correlation between fear-based disorders and GAD
The genetic correlation between fear and GAD was high (rg= 0.85-0.96) and significantly
different from 0 when analysing both GWAS phenotypes (fearspecific–GADspecific and fearanx-dep
GADanx-dep) in both GWAS meta-analyses datasets (full meta-analysis and GLAD+ and UKB
meta-analysis excluding depression-enriched QIMR dataset). The genetic correlation between
fear and GAD was significantly less than 1 when controls were screened specifically and when
the QIMR dataset was excluded from the meta-analysis. All other fear–GAD genetic
correlations were not significantly different from 1 (Table 4; Bonferroni-corrected significance
threshold P ≤ 0.0125).

Page 70
69
Table 4: Genetic correlations (rg ) between fear-based disorder and GAD phenotypes
Datasets Control criteria
rg
se
Z
P (0)
P (1)
GLAD+ &
UKB
Specific
0.85
0.05
15.63
4.90E-55
4.57E-03
Any anx-dep
0.92
0.03
27.52
9.65E-167
0.0215
QIMR,
GLAD+ &
UKB
Specific
0.95
0.03
28.77
4.77E-182
0.1282
Any anx-dep
0.97
0.02
47.03
0.00E+00
0.1808
Genetic correlations estimated using LDSC regression. P (0) - P-value for test of genetic
correlation different from 0. P (1) - P-value for test of genetic correlation different from 1.
Genetic correlations with external phenotypes
We calculated genetic correlations between fear and GAD with 345 external phenotypes. Only
fearspecific and GADspecific in the GWAS meta-analysis excluding the depression-enriched QIMR
dataset were tested for genetic correlations with external phenotypes (as the fear–GAD
genetic correlation was significantly different from 1). There was no statistically significant
difference in genetic correlations with most external traits across fearspecific and GADspecific
(Bonferroni-corrected significance threshold P 1.45 � 10-4; Figure 2; left panel). Exceptions
were observed for educational attainment, general cognitive ability, which showed significantly
stronger negative genetic correlations with fearspecific than with GADspecific. Coronary artery
disease also showed a significantly higher positive genetic correlation with fearspecific than with
GADspecific. Conversely, a diagnosis of any bipolar disorder (type I and type II), bipolar disorder
type I, anorexia nervosa, and neuroticism showed significantly stronger positive genetic
correlationswith GADspecific than with fearspecific (Figure 2; right panel). Full genetic correlation
results with external phenotypes are shown in Supplementary Table 9.

Page 71
70
Figure 2: Genetic correlations between anxiety disorder phenotypes and external traits estimated in LDSC regression.
Fear-based disorder GWAS and GAD GWAS results from phenotypes with specifically screened controls, meta-analysed across the UKB and GLAD+ datasets.
Genetic correlations with 345 phenotypes were tested (Bonferroni-corrected significance threshold P ≤ 1.45 � 10-4 ). Only traits with genetic correlation P-values
significantly different from 0 with at least one of the anxiety disorder phenotypes are shown (after Bonferonni correction for multiple testing). Full results are shown in
Supplementary Table 9. External phenotypes were tested for significantly different genetic correlations with fear-based disorders versus GAD using a block
jackknife (Bonferroni correction for significance [P ≤ 1.45 � 10-4]). Left panel: phenotypes with no statistically significant different genetic correlations between fear-
based disorders and GAD, though in the lower two segments, there was a significant association with one trait and not the other. Right panel: phenotypes with
statistically significant different genetic correlations between fear-based disorders and GAD. Bars represent standard errors. ADHD = attention deficit hyperactivity
disorder; ASD = autism spectrum disorder; BIP = bipolar disorder; GAD = generalised anxiety disorder; MDD = major depressive disorder; OCD = obsessive-
compulsive disorder; PGC = Psychiatric Genomics Consortium; PTSD = post-traumatic stress disorder; SCZ = schizophrenia; UKB = UK Biobank.
.8 1.0
#
#
#
#
#
#
#
#
#
GAD & fear rg significantly different from each other
Significantly stronger rg with fear
Significantly stronger rg with GAD
−0.6−0.4−0.20.0 0.2 0.4 0.6 0.8 1.0
Educational Attainment
College/university degree attainment
General cognitive ability
Coronary Artery Disease
Anorexia nervosa
Bipolar disorder type I
Bipolar disorder type I & II
Cross disorders (ADHD, ASD, BIP, SCZ)
Neuroticism UKB
Genetic correlation rg
#
#
Fear−based disorders: significant
GAD: significant
Fear−based disorders: non−significant
GAD: non−significant
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
#
GAD & fear rg not significantly different from each other
Both significantly different from 0
Only fear signif.
Only GAD signif.
−0.6−0.4−0.2 0.0 0.2 0.4 0.6 0.8 1.0
Ever smoker
Automobile speeding
ADHD
Schizophrenia
Insomnia
Childhood maltreatment
Self−rated health
Bipolar disorder type II
Subjective well−being
Self−reported tiredness
PTSD symptoms
GAD symptoms
Neuroticism, no psychiatric illness
MDD (PGC2, no 23andme, no UKB)
Depressive symptoms
Cross 12 psychiatric disorders
MDD (PGC2 + 23andme + UKB)
Lifetime anxiety
Body fat percentage
Verbal−numerical reasoning
Father's age at death
Mother's age at death
Age first birth
Undersleeper
Household income
Problematic consequences of drinking
Autism spectrum disorder
OCD
Alcohol dependence
Genetic correlation rg
College
Cross disor

Page 72
71
Discussion
This is the first study to report genome-wide significant loci (three in total) associated with any
lifetime fear-based disorder diagnosis, hereafter referred to as fear. In addition, we identified
four genome-wide significant loci for lifetime GAD diagnosis, previously identified in other
GWAS of anxiety15,48 and distress-related traits49–51. The increase in power compared to
previous GWAS of fear19 was achieved by combining detailed and brief diagnostic measures.
In agreement with twin studies, we found that fear and GAD were highly genetically correlated
with each other and with depression9,11,12. When we excluded the depression-enriched dataset
and only screened controls for the specific anxiety disorder being analysed, genetic
correlations between these disorders were reduced, suggesting some genetic specificity
between them. Differences in common genetic variant-based genetic correlations between
fear and GAD with broad domains of other complex traits were explored. Compared with GAD,
fear showed significantly stronger negative genetic correlations with general cognitive ability
and educational attainment and stronger positive genetic correlations with coronary artery
disease. Conversely, neuroticism, bipolar disorder type I and anorexia nervosa were among
the traits with significantly higher positive genetic correlations with GAD than fear.
We found independent loci and gene-level associations that support previous GWAS findings
of other phenotypes and some novel associations. We identified a novel variant associated
with fear that has not been previously reported in GWASs of complex traits. The nearest gene
to the intergenic variant is the uncharacterised, non-coding gene CTB-57H20.1. Other loci and
gene-level associations with fear that we identified have been previously implicated in GWAS
of educational attainment52, general cognitive ability53, insomnia54 and broad depression49,
including sorting nexin 29. The sorting nexin family of proteins is thought to play a role in
neuronal function, synaptic plasticity, learning, and memory55. One locus associated with fear
was previously reported in a worry-neuroticism GWAS in the UK Biobank50. No fear loci or
gene-level associations overlapped with our GAD results. As GWASs of specific anxiety
disorders improve in power, we expect this overlap to increase.
Some of our loci and gene-level associations with GAD were reported in previous UK Biobank
GWASs of lifetime anxiety and GAD symptoms15,48, which is expected given the sample
overlap. Loci and gene-level associations with GAD were consistently replicated in previous
GWASs of other distress-related phenotypes, with all four loci previously identified in
depression and neuroticism49–51. This includes the SORCS3 gene, which is highly expressed
in the CA1 hippocampus region and has been implicated in aversive memory extinction in
mice56. SORCS3 was also associated with anxiety-like behaviour in mice, mediated by
changes in gut microbiome measures suggesting the role of the gut-brain axis57. Our finding

Page 73
72
warrants further animal modelling and functional genomic interrogation of the role of SORCS3
in distress liability and translational potential.
The high genetic correlations we observed between fear, GAD, and depression are consistent
with previous twin studies9,11,12. Our results are compatible with our previous preliminary
GWAS in the UK Biobank19 but add further detail to the genetic structure of fear and GAD by
assessing a broad range of other complex traits. We verified that screening controls
specifically for the disorder grouping being analysed improved our ability to distinguish genetic
differences between highly genetically correlated disorders (e.g. controls in the fear GWAS
were only screened for any fear-based disorder). We found evidence of some genetic
differences between fear and GAD once we screened controls specifically and after excluding
the depression-ascertained QIMR dataset. This highlights the necessity for careful
consideration of how to screen controls when attempting to identify disorder-specific genetics.
Genetic correlations with depression were high and not significantly different between fear
and GAD, supporting a common genetic liability factor shared among these disorders9,11,12,58.
A common liability factor is often conceptualised as being driven by shared negative affectivity,
which is also well captured by measures of the personality trait neuroticism22,59. We found
neuroticism had a significantly stronger genetic correlation with GAD than fear. This is in line
with twin studies that reported genetic influences on GAD (and depression) were more core
to a higher-order dimension of genetic liability to negative affectivity than fear60. General
negative affectivity also influences fear-based disorders but is less central to them; instead,
they are characterised by more specific elements of acute fearful and physiological
hyperarousal61. Our findings partially support a distress-fear genetic distinction. However, we
found stronger evidence for an overarching genetic liability to internalising that drives
comorbidity across anxiety disorders and depression.
Compared with GAD, fear had stronger negative genetic correlations with educational
attainment and general cognitive ability and a higher positive genetic correlation with coronary
artery disease (heart disease). These correlations were non-significant with GAD. The specific
genetic relationship observed between fear and cognitive-related traits may reflect shared
cognitive mechanisms, such as those involved in learning, that are less central to GAD.
Associative learning is a key mechanism that distinguishes fear-based disorders from GAD6.
Our finding that heart disease is uniquely genetically correlated with fear may be explained by
genetics playing a bigger part in driving comorbidity between heart disease and fear than with
GAD. Core symptoms of fear-based disorders, including experiencing phobic fears and
somatic arousal, are linked to a higher risk of heart disease and cardiac fatality 62–65. Our
findings align with a phenotypic study that revealed fear-based disorders were stronger

Page 74
73
predictors of heart disease development than distress disorders66. The earlier age of onset of
chronically experienced fear-based disorders compared to distress may give rise to longer-
term exposure to mechanisms associated with heart disease risk1,66,67. Overlapping genetics
with heart disease may underpin shared physiological mechanisms in inflammatory pathways
or autonomic dysfunction68.
GAD showed significantly stronger positive genetic correlations than fear with a diagnosis of
any bipolar disorder (type I or II). Bipolar type II disorder involves episodes of hypomania and
is more similar to depression, whereas bipolar type I disorder is characterised by more severe
manic episodes4. When we analysed bipolar disorder I and II separately, we found GAD and
fear had no significantly different genetic correlation with bipolar II, whereas bipolar I was
significantly more positively genetically correlated with GAD. This indicates that bipolar II
drives down the genetic correlations with fear when analysed together with bipolar I. This
aligns with previous studies that found bipolar I had a lower genetic correlation with depression
than bipolar II and was more genetically similar to rarer psychiatric illnesses, including
schizophrenia69,70. Genetic correlation analyses of the mood disorder spectrum reflect a
genetic continuum from manic to depressive clusters. Bipolar I sits on one end of the spectrum,
and depressive disorders on the other, with bipolar type II connecting the two disorders71. Our
novel finding adds further fine-grained detail to this genetic structure by highlighting
heterogeneity at the internalising pole of the genetic continuum. On the opposite end of the
spectrum to bipolar type I lies fear-based disorders, with distress disorders (GAD and
depression) sitting between bipolar and fear-based disorders.
We found that fear had a significantly weaker genetic correlation with anorexia nervosa (~0.10)
than GAD (~0.30). This is somewhat unexpected given that fear-based disorders have similar
rates of comorbidity with anorexia nervosa compared to GAD, thought to be driven by an
overlap in genetic susceptibility and fear-conditioning mechanisms72–74. Few studies have
assessed genetic correlations specifically between anorexia nervosa and the individual
anxiety disorder subtypes, with analyses restricted to GAD75. However, the genetic overlap
between other eating disorder phenotypes and fear-based disorders has been reported76,77.
One twin study found that a broad eating disorder phenotype had moderate genetic
correlations with panic disorder (0.62) and GAD (0.51) and a slightly lower genetic correlation
with specific phobias (0.40), agoraphobia (0.38), and social anxiety disorder (0.33)77. Thus,
further research is required to establish whether genetic correlations with anorexia nervosa
differ between individual fear-based disorders. There may also be shared loci that affect traits
in different directions, making us unable to detect a global genetic correlation. Studies
conducting local genetic correlation analyses may identify specific shared regions of the

Page 75
74
genome supporting shared mechanisms that link fear-based disorders with anorexia
nervosa78,79.
Our study is the first to identify fear-based disorder-associated genetic loci. However, findings
should be considered in light of limitations that impacted our ability to detect fear-specific
genetic influences. First, we were underpowered to assess the fear-based disorders
individually. Some twin studies suggest the proportion of disorder-specific genetic influences
on individual fear-based disorders varies11,12, with specific phobia being less genetically similar
to GAD than panic disorder and agoraphobia11. Second, results may have differed if we had
sufficient sample sizes to only use panic disorder diagnosis instead of combining it with panic
attack measures. Panic attacks are transdiagnostic and can be included as a specifier for a
range of DSM-5 psychiatric disorders4. Using self-reports of panic attacks may have elevated
the genetic correlations between fear and other psychiatric disorders. However,
epidemiological estimates of lifetime panic disorder comorbidity with other mental disorders
are also high (80%)80. Third, specifically screening controls for a certain anxiety disorder was
not possible in the QIMR data as we were limited to using a single-item brief measure of any
anxiety disorder. Fourth, although a strength of our study was incorporating detailed measures
of anxiety disorders, we could not compare genetic differences between brief versus detailed
diagnostic measures. Studies suggest that more detailed measures increase genetic
specificity25. Combining detailed with brief measures may have limited genetic specificity and
elevated the genetic correlation between fear, GAD, and related disorders such as depression.
Finally, we could not account for the high comorbidity observed across anxiety disorders and
depression. We previously found that cases rarely met criteria for only one anxiety disorder
and no comorbid MDD (5%) in the GLAD+ dataset81. Thus, ascertaining data for well-powered
GWAS of a single anxiety disorder without comorbidities will be challenging and are less
representative of the broader lived experience of these disorders. We did not include heritable
comorbid traits as GWAS covariates as this can bias results82. Future studies with well-
powered GWAS of anxiety subtypes could discern subtype-specific genetics through
multivariate genomic structural equation modelling83. As individual-level methods advance84,85,
this will enable testing genetic covariances across comorbid disorders and comparing
measures in smaller samples than is required for GWAS summary-level modelling.
In summary, our results add further evidence that fear-based disorders and GAD share a high
proportion of genetic liability, as well as with depression, while also revealing some genetic
differences between them. We identified differences in the genetic relationship between fear
and GAD, including those with neuroticism, bipolar type I, anorexia nervosa, general cognitive
ability, and coronary artery disease. Our findings add more fine-grained detail to the proposed

Page 76
75
hierarchical structure of internalising disorders and partially support fear-based disorders and
GAD as separate subfactors of distress and fear. Quantitatively-based dimensional modelling
of shared and distinct distress and fear symptoms on the genomic level would further complete
this hierarchical structure22. The growth of datasets with detailed phenotyping of all anxiety
disorders, such as those used in this study, will be key for further identifying subtype-specific
and transdiagnostic genetic factors of the full anxiety disorder spectrum.

Page 77
76
References
1. Kessler, R. C. et al. Lifetime prevalence and age-of-onset distributions of DSM-IV
disorders in the National Comorbidity Survey Replication. Arch. Gen. Psychiatry 62,
593–602 (2005).
2. Somers, J. M., Goldner, E. M., Waraich, P. & Hsu, L. Prevalence and incidence
studies of anxiety disorders: a systematic review of the literature. Can. J. Psychiatry
51, 100–113 (2006).
3. Craske, M. G. et al. Anxiety disorders. Nat Rev Dis Primers 3, 17024 (2017).
4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental
Disorders (DSM-5�). (American Psychiatric Pub, 2013).
5. Kessler, R. C. The global burden of anxiety and mood disorders: putting the
European Study of the Epidemiology of Mental Disorders (ESEMeD) findings into
perspective. J. Clin. Psychiatry 68 Suppl 2, 10–19 (2007).
6. Craske, M. G. & Waters, A. M. Panic Disorder, Phobias, and Generalized Anxiety
Disorder. Annual Review of Clinical Psychology vol. 1 197–225 Preprint at
https://doi.org/10.1146/annurev.clinpsy.1.102803.143857 (2005).
7. Polderman, T. J. C. et al. Meta-analysis of the heritability of human traits based on
fifty years of twin studies. Nat. Genet. 47, 702–709 (2015).
8. Ask, H. et al. Genetic contributions to anxiety disorders: where we are and where we
are heading. Psychol. Med. 51, 2231–2246 (2021).
9. Waszczuk, M. A., Zavos, H. M. S., Gregory, A. M. & Eley, T. C. The phenotypic and
genetic structure of depression and anxiety disorder symptoms in childhood,
adolescence, and young adulthood. JAMA Psychiatry 71, 905–916 (2014).
10. Roberson-Nay, R., Eaves, L. J., Hettema, J. M., Kendler, K. S. & Silberg, J. L.
Childhood separation anxiety disorder and adult onset panic attacks share a common
genetic diathesis. Depress. Anxiety 29, 320–327 (2012).
11. Hettema, J. M., Prescott, C. A., Myers, J. M., Neale, M. C. & Kendler, K. S. The
structure of genetic and environmental risk factors for anxiety disorders in men and
women. Arch. Gen. Psychiatry 62, 182–189 (2005).
12. Tambs, K. et al. Structure of genetic and environmental risk factors for dimensional
representations of DSM–IV anxiety disorders. Br. J. Psychiatry 195, 301–307 (2009).
13. Kendler, K. S., Prescott, C. A., Myers, J. & Neale, M. C. The structure of genetic and
environmental risk factors for common psychiatric and substance use disorders in
men and women. Arch. Gen. Psychiatry 60, 929–937 (2003).
14. Smoller, J. W. Anxiety Genetics Goes Genomic. The American journal of psychiatry
vol. 177 190–194 (2020).
15. Purves, K. L. et al. A major role for common genetic variation in anxiety disorders.
Mol. Psychiatry 25, 3292–3303 (2020).
16. Levey, D. F. et al. Reproducible Genetic Risk Loci for Anxiety: Results From
∼200,000 Participants in the Million Veteran Program. Am. J. Psychiatry 177, 223–
232 (2020).
17. Meier, S. M. et al. Genetic Variants Associated With Anxiety and Stress-Related
Disorders: A Genome-Wide Association Study and Mouse-Model Study. JAMA
Psychiatry (2019) doi:10.1001/jamapsychiatry.2019.1119.
18. Wendt, F. R. et al. Using phenotype risk scores to enhance gene discovery for
generalized anxiety disorder and posttraumatic stress disorder. Mol. Psychiatry
(2022) doi:10.1038/s41380-022-01469-y.
19. Morneau-Vaillancourt, G. et al. The genetic and environmental hierarchical structure
of anxiety and depression in the UK Biobank. Depress. Anxiety 37, 512–520 (2020).
20. Deckert, J. et al. GLRB allelic variation associated with agoraphobic cognitions,
increased startle response and fear network activation: a potential neurogenetic
pathway to panic disorder. Mol. Psychiatry 22, 1431–1439 (2017).

Page 78
77
21. Erhardt, A. et al. TMEM132D, a new candidate for anxiety phenotypes: evidence
from human and mouse studies. Mol. Psychiatry 16, 647–663 (2011).
22. Watson, D. et al. Validity and utility of Hierarchical Taxonomy of Psychopathology
(HiTOP): III. Emotional dysfunction superspectrum. World Psychiatry 21, 26–54
(2022).
23. Toft, T. et al. Mental disorders in primary care: prevalence and co-morbidity among
disorders. results from the functional illness in primary care (FIP) study. Psychol.
Med. 35, 1175–1184 (2005).
24. Kendler, K. S., Chatzinakos, C. & Bacanu, S.-A. The impact on estimations of genetic
correlations by the use of super-normal, unscreened, and family-history screened
controls in genome wide case-control studies. Genet. Epidemiol. 44, 283–289 (2020).
25. Cai, N., Choi, K. W. & Fried, E. I. Reviewing the genetics of heterogeneity in
depression: operationalizations, manifestations and etiologies. Hum. Mol. Genet. 29,
R10–R18 (2020).
26. Flint, J. The genetic basis of major depressive disorder. Mol. Psychiatry (2023)
doi:10.1038/s41380-023-01957-9.
27. Davies, M. R. et al. Comparison of symptom-based versus self-reported diagnostic
measures of anxiety and depression disorders in the GLAD and COPING cohorts. J.
Anxiety Disord. 85, 102491 (2022).
28. Mitchell, B. L. et al. The Australian Genetics of Depression Study: New Risk Loci and
Dissecting Heterogeneity Between Subtypes. Biol. Psychiatry (2021)
doi:10.1016/j.biopsych.2021.10.021.
29. Davis, K. A. S. et al. Mental health in UK Biobank - development, implementation and
results from an online questionnaire completed by 157 366 participants: a reanalysis.
BJPsych Open 6, e18 (2020).
30. Gigantesco, A. & Morosini, P. Development, reliability and factor analysis of a self-
administered questionnaire which originates from the World Health Organization’s
Composite International Diagnostic Interview – Short Form (CIDI-SF) for assessing
mental disorders. Clinical Practice and Epidemiology in Mental Health vol. 4 8
Preprint at https://doi.org/10.1186/1745-0179-4-8 (2008).
31. Byrne, E. M. et al. Cohort profile: the Australian genetics of depression study. BMJ
Open 10, e032580 (2020).
32. Davies, M. R. et al. The Genetic Links to Anxiety and Depression (GLAD) Study:
Online recruitment into the largest recontactable study of depression and anxiety.
Behav. Res. Ther. 123, 103503 (2019).
33. Young, K. S. et al. Depression, anxiety and PTSD symptoms before and during the
COVID-19 pandemic in the UK. Psychol. Med. 1–14 (2022).
34. Olsen, C. M. et al. Cohort profile: the QSkin Sun and Health Study. Int. J. Epidemiol.
41, 929–929i (2012).
35. Sudlow, C. et al. UK biobank: an open access resource for identifying the causes of a
wide range of complex diseases of middle and old age. PLoS Med. 12, e1001779
(2015).
36. Mbatchou, J. et al. Computationally efficient whole-genome regression for
quantitative and binary traits. Nat. Genet. 53, 1097–1103 (2021).
37. Zhou, W. et al. Efficiently controlling for case-control imbalance and sample
relatedness in large-scale genetic association studies. Nat. Genet. 50, 1335–1341
(2018).
38. Willer, C. J., Li, Y. & Abecasis, G. R. METAL: fast and efficient meta-analysis of
genomewide association scans. Bioinformatics 26, 2190–2191 (2010).
39. Watanabe, K., Taskesen, E., van Bochoven, A. & Posthuma, D. Functional mapping
and annotation of genetic associations with FUMA. Nat. Commun. 8, 1826 (2017).
40. de Leeuw, C. A., Mooij, J. M., Heskes, T. & Posthuma, D. MAGMA: generalized
gene-set analysis of GWAS data. PLoS Comput. Biol. 11, e1004219 (2015).

Page 79
78
41. Bulik-Sullivan, B. K. et al. LD Score regression distinguishes confounding from
polygenicity in genome-wide association studies. Nature Genetics vol. 47 291–295
Preprint at https://doi.org/10.1038/ng.3211 (2015).
42. Bulik-Sullivan, B. et al. An atlas of genetic correlations across human diseases and
traits. Nat. Genet. 47, 1236–1241 (2015).
43. H�bel, C. et al. Genomics of body fat percentage may contribute to sex bias in
anorexia nervosa. Am. J. Med. Genet. B Neuropsychiatr. Genet. 180, 428–438
(2019).
44. Loh, P.-R., Kichaev, G., Gazal, S., Schoech, A. P. & Price, A. L. Mixed-model
association for biobank-scale datasets. Nat. Genet. 50, 906–908 (2018).
45. Davies, H. L. et al. Risk and protective factors for new-onset binge eating, low
weight, and self-harm symptoms in> 35,000 individuals in the UK during the COVID-
19 pandemic. Int. J. Eat. Disord. (2022).
46. Bandelow, B. & Michaelis, S. Epidemiology of anxiety disorders in the 21st century.
Dialogues Clin. Neurosci. 17, 327–335 (2015).
47. Leray, E. et al. Prevalence, characteristics and comorbidities of anxiety disorders in
France: Results from the ‘Mental Health in General Population’ Survey (MHGP). Eur.
Psychiatry 26, 339–345 (2011).
48. Thorp, J. G. et al. Symptom-level modelling unravels the shared genetic architecture
of anxiety and depression. Nat Hum Behav 5, 1432–1442 (2021).
49. Howard, D. M. et al. Genome-wide meta-analysis of depression identifies 102
independent variants and highlights the importance of the prefrontal brain regions.
Nat. Neurosci. 22, 343–352 (2019).
50. Nagel, M. et al. Meta-analysis of genome-wide association studies for neuroticism in
449,484 individuals identifies novel genetic loci and pathways. Nat. Genet. 50, 920–
927 (2018).
51. Wray, N. R. et al. Genome-wide association analyses identify 44 risk variants and
refine the genetic architecture of major depression. Nat. Genet. 50, 668–681 (2018).
52. Okbay, A. et al. Polygenic prediction of educational attainment within and between
families from genome-wide association analyses in 3 million individuals. Nat. Genet.
54, 437–449 (2022).
53. Davies, G. et al. Study of 300,486 individuals identifies 148 independent genetic loci
influencing general cognitive function. Nat. Commun. 9, 2098 (2018).
54. Watanabe, K. et al. Genome-wide meta-analysis of insomnia prioritizes genes
associated with metabolic and psychiatric pathways. Nat. Genet. 54, 1125–1132
(2022).
55. Vieira, N., Rito, T., Correia-Neves, M. & Sousa, N. Sorting Out Sorting Nexins
Functions in the Nervous System in Health and Disease. Mol. Neurobiol. 58, 4070–
4106 (2021).
56. Breiderhoff, T. et al. Sortilin-related receptor SORCS3 is a postsynaptic modulator of
synaptic depression and fear extinction. PLoS One 8, e75006 (2013).
57. Jin, X. et al. Gut microbiome partially mediates and coordinates the effects of
genetics on anxiety-like behavior in Collaborative Cross mice. Sci. Rep. 11, 270
(2021).
58. Mosing, M. A. et al. Genetic and environmental influences on the co-morbidity
between depression, panic disorder, agoraphobia, and social phobia: a twin study.
Depress. Anxiety 26, 1004–1011 (2009).
59. Hettema, J. M., Neale, M. C., Myers, J. M., Prescott, C. A. & Kendler, K. S. A
population-based twin study of the relationship between neuroticism and internalizing
disorders. Am. J. Psychiatry 163, 857–864 (2006).
60. Kendler, K. S. & Myers, J. The boundaries of the internalizing and externalizing
genetic spectra in men and women. Psychol. Med. 44, 647–655 (2014).
61. Watson, D. Rethinking the mood and anxiety disorders: a quantitative hierarchical
model for DSM-V. J. Abnorm. Psychol. 114, 522–536 (2005).

Page 80
79
62. Watkins, L. L. et al. Phobic anxiety and increased risk of mortality in coronary heart
disease. Psychosom. Med. 72, 664–671 (2010).
63. Albert, C. M., Chae, C. U., Rexrode, K. M., Manson, J. E. & Kawachi, I. Phobic
anxiety and risk of coronary heart disease and sudden cardiac death among women.
Circulation 111, 480–487 (2005).
64. Nabi, H. et al. Psychological and somatic symptoms of anxiety and risk of coronary
heart disease: the health and social support prospective cohort study. Biol.
Psychiatry 67, 378–385 (2010).
65. Roest, A. M., Heideveld, A., Martens, E. J., de Jonge, P. & Denollet, J. Symptom
dimensions of anxiety following myocardial infarction: associations with depressive
symptoms and prognosis. Health Psychol. 33, 1468–1476 (2014).
66. Roest, A. M. et al. Fear and distress disorders as predictors of heart disease: A
temporal perspective. J. Psychosom. Res. 96, 67–75 (2017).
67. Lijster, J. M. de et al. The Age of Onset of Anxiety Disorders. Can. J. Psychiatry 62,
237–246 (2017).
68. Celano, C. M., Daunis, D. J., Lokko, H. N., Campbell, K. A. & Huffman, J. C. Anxiety
Disorders and Cardiovascular Disease. Curr. Psychiatry Rep. 18, 101 (2016).
69. Mallard, T. T. et al. Multivariate GWAS of psychiatric disorders and their cardinal
symptoms reveal two dimensions of cross-cutting genetic liabilities. Cell Genom 2,
(2022).
70. Mullins, N. et al. Genome-wide association study of more than 40,000 bipolar
disorder cases provides new insights into the underlying biology. Nat. Genet. 53,
817–829 (2021).
71. Coleman, J. R. I. et al. The Genetics of the Mood Disorder Spectrum: Genome-wide
Association Analyses of More Than 185,000 Cases and 439,000 Controls. Biol.
Psychiatry (2019) doi:10.1016/j.biopsych.2019.10.015.
72. Meier, S. M. et al. Diagnosed Anxiety Disorders and the Risk of Subsequent
Anorexia Nervosa: A Danish Population Register Study. Eur. Eat. Disord. Rev. 23,
524–530 (2015).
73. Kaye, W. H., Bulik, C. M., Thornton, L., Barbarich, N. & Masters, K. Comorbidity of
anxiety disorders with anorexia and bulimia nervosa. Am. J. Psychiatry 161, 2215–
2221 (2004).
74. Godart, N. T., Flament, M. F., Perdereau, F. & Jeammet, P. Comorbidity between
eating disorders and anxiety disorders: a review. Int. J. Eat. Disord. 32, 253–270
(2002).
75. Dellava, J. E., Kendler, K. S. & Neale, M. C. Generalized anxiety disorder and
anorexia nervosa: evidence of shared genetic variation. Depress. Anxiety 28, 728–
733 (2011).
76. Kendler, K. S. et al. The structure of the genetic and environmental risk factors for six
major psychiatric disorders in women. Phobia, generalized anxiety disorder, panic
disorder, bulimia, major depression, and alcoholism. Arch. Gen. Psychiatry 52, 374–
383 (1995).
77. Kendler, K. S. et al. The structure of genetic and environmental risk factors for
syndromal and subsyndromal common DSM-IV axis I and all axis II disorders. Am. J.
Psychiatry 168, 29–39 (2011).
78. Werme, J., van der Sluis, S. & de Leeuw, C. A. LAVA: An integrated framework for
local genetic correlation analysis. bioRxiv 2020.12.31.424652 (2021)
doi:10.1101/2020.12.31.424652.
79. Ronald, A. & Pain, O. A Revolution Is Brewing in How We Understand the Shared
Genetic Causes of Psychiatric Disorders. The American journal of psychiatry vol. 179
791–793 (2022).
80. de Jonge, P., Roest, A. M., Lim, C. C. W., Levinson, D. & Scott, K. M. Panic Disorder
and Panic Attacks. in Mental Disorders Around the World: Facts and Figures from the
WHO World Mental Health Surveys 93–105 (Cambridge University Press, 2018).

Page 81
80
81. Davies, M. R. et al. Factors associated with anxiety disorder comorbidity. J. Affect.
Disord. 323, 280–291 (2023).
82. Aschard, H., Vilhj�lmsson, B. J., Joshi, A. D., Price, A. L. & Kraft, P. Adjusting for
heritable covariates can bias effect estimates in genome-wide association studies.
Am. J. Hum. Genet. 96, 329–339 (2015).
83. Grotzinger, A. D. et al. Genomic structural equation modelling provides insights into
the multivariate genetic architecture of complex traits. Nat Hum Behav 3, 513–525
(2019).
84. de Hoyos, L. et al. Structural models of genome-wide covariance identify multiple
common dimensions in autism. bioRxiv (2022) doi:10.1101/2022.10.21.22281213.
85. Pritikin, J. N., Neale, M. C., Prom-Wormley, E. C., Clark, S. L. & Verhulst, B. GW-
SEM 2.0: Efficient, Flexible, and Accessible Multivariate GWAS. Behav. Genet. 51,
343–357 (2021).

Page 82
81
Chapter 3. Genetic decomposition of the heritable
component of reported childhood maltreatment
This chapter is presented as a manuscript of an accepted paper in Biological Psychiatry:
Global Open Science. It is an exact copy of this manuscript. Supplementary materials for this
chapter, as detailed in the text, are included in Appendix B.
In press citation: ter Kuile, A. R., H�bel, C., Cheesman, R., Coleman, J. R. I., Peel, A. J.,
Levey, D. F., Stein, M. B., Gelernter, J., Rayner, C., Eley, T. C., & Breen, G. (In press). Genetic
decomposition of the heritable component of reported childhood maltreatment. Biological
Psychiatry Global Open Science. https://doi.org/10.1016/j.bpsgos.2023.03.003
Authors: Abigail R. ter Kuile1,2, Christopher H�bel1,2,3, Rosa Cheesman8, Jonathan R. I.
Coleman1,2, Alicia J. Peel1, Daniel F. Levey4,5, Murray B. Stein6,7, Joel Gelernter4,5,
Christopher Rayner1, Thalia C. Eley1,2, Gerome Breen1,2
1. Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry,
Psychology & Neuroscience, King’s College London, UK
2. National Institute for Health and Care Research (NIHR) Maudsley Biomedical
Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
3. National Centre for Register-based Research, Aarhus Business and Social Sciences,
Aarhus University, Aarhus, Denmark
4. Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
5. Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
6. Department of Psychiatry and School of Public Health, University of California San
Diego, La Jolla, CA, USA
7. VA San Diego Healthcare System, San DIego, CA, USA
8. PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo,
Norway

Page 83
82
Abstract
Background: Decades of research have shown that environmental exposures, including self-
reports of trauma, are partly heritable. Heritable characteristics may influence exposure to and
interpretations of environmental factors. Identifying heritable factors associated with self-
reported trauma could improve our understanding of vulnerability to exposure and the
interpretation of life events.
Methods: We used genome-wide association study summary statistics of childhood
maltreatment, defined as reporting of abuse (emotional, sexual and physical) and neglect
(emotional and physical) (N=185,414). We calculated genetic correlations (rg) between
reported childhood maltreatment and 576 traits to identify phenotypes that might explain the
heritability of reported childhood maltreatment, retaining those with |rg|>0.25. We specified
multiple regression models using genomic structural equation modelling to detect residual
genetic variance in childhood maltreatment after accounting for genetically correlated traits.
Results: In two separate models, the shared genetic component of twelve health and
behavioural traits and seven psychiatric disorders accounted for 59% and 56% of heritability
due to common genetic variants (h2
SNP) of childhood maltreatment, respectively. Genetic
influences on the h2
SNP of childhood maltreatment were generally accounted for by a shared
genetic component across traits. The exceptions to this were general risk tolerance, subjective
well-being, post-traumatic stress disorder and autism spectrum disorder, identified as
independent contributors to its h2
SNP. These four traits alone were sufficient to explain 58% of
the h2
SNP of childhood maltreatment.
Conclusions: We identified putative traits that reflect the h2
SNP of childhood maltreatment.
Elucidating the mechanisms underlying these associations may improve trauma prevention
and posttraumatic intervention strategies.

Page 84
83
Introduction
Traumatic events, namely those perceived as physically or emotionally threatening and
violating, are associated with various adverse outcomes, including psychopathology1–4.
Decades of behavioural genetics research has shown that reported trauma exposures, like
many environmental measures and behavioural traits, are partly heritable5,6. Twin studies
estimate 6-62% of the variance in reporting different types of trauma is attributable to
genetics7–12. Interpersonal assaultive traumas (e.g., physical and sexual assault) have higher
heritability than non-interpersonal or non-assaultive traumas (e.g., accidents)7,9,13. In relation
to these observations, stressful life events (SLEs) dependent on one’s behaviour (e.g., fights)
are more heritable than those that are independent (e.g., natural disasters), with the latter
occurring more often due to chance14. However, being at higher genetic risk for reported
trauma does not signify that an individual is genetically predestined to experience trauma.
Furthermore, a large proportion of the total phenotypic variability of reported trauma is not
attributable to genetics. The environment itself may be harmful, or a perpetrator may exploit
those in vulnerable circumstances11,15. However, environmental risk factors are generally
unstable, idiosyncratic, and thus, unpredictable and challenging to examine16. Exploring traits
genetically related to reported trauma in different environmental contexts may provide a
framework for social research to help determine trauma risk factors and protect vulnerable
individuals6.
Heritable behavioural characteristics may contribute to the likelihood of experiencing certain
events. Personality traits, such as openness to experience and antisocial behaviour, are
phenotypically and genetically correlated with reporting interpersonal assaultive trauma17.
Such partially heritable characteristics may contribute to the heritability of reported trauma
through gene-environment correlation (rGE), whereby the environment reflects an individual’s
genetic propensities via three different processes18. Passive rGE occurs when a relative’s
genotype, such as parental genetic variation contributing to risk-taking behaviours, shapes the
child’s environment and potentially creates an unsafe home19,20. The environment that the
parent creates and the parental genotype are correlated as the child receives both from their
biological parents. Thus, parental environmental effects may be captured in genetic analyses
of offspring traits18. Evocative rGE arises when an individual’s genotype shapes how others
engage with them. For example, a child’s behavioural difficulties may evoke verbal and
physical discipline due to the carer’s expectations of how a child should behave18. Active rGE
involves an individual’s genetic disposition to, for example, risk-taking modifying and selecting
their environment18,21, leading to differing risks of exposure to adverse environments.

Page 85
84
Correlations between genetic factors and retrospective reports of trauma may also, in part, be
driven by heritable characteristics influencing the subjective interpretation, willingness to
disclose and recollection of events22,23. Genetic research has largely relied on retrospective
self-reports of trauma exposure which may be more susceptible to genetically influenced
perceptions and recollection of events, as opposed to more objective measures prospectively
recorded closer to the time of exposure (e.g., court records, caregiver reports)23. Memory,
emotional regulation and interpretation biases are partly heritable24–27 and are associated with
retrospective reporting of trauma in early life28. Individual differences in subjective experiences
are partly influenced by genetics29,30. Subjective appraisal of trauma is important for
posttraumatic psychopathology, which is more strongly associated with retrospective self-
reports of trauma than objective court records31. Individual, partially heritable differences in
personality traits such as neuroticism and agreeableness may explain the discrepancy
between retrospective and prospective measures of trauma31–33. Furthermore, the consistency
and frequency of self-reports are impacted by individual factors involved in the willingness to
disclose a traumatic event, such as perception of stigma, fear of negative consequences, or
pre-existing relationships with the perpetrator34–37. Lack of disclosure is a barrier to therapeutic
and legal interventions36. Thus, a better understanding of the heritable factors that impact the
retrospective report of trauma experiences could help improve posttraumatic support.
In sum, the influences on retrospectively reported trauma are complex and difficult to
disentangle. A range of heritable traits may be involved. Heritability and genetic correlations
between traits can be estimated using genome-wide association study (GWAS) summary
statistics38. The proportion of heritability explained by common genetic variants (h2
SNP) ranges
from 6-9% for reported interpersonal trauma during childhood21,39 to 18% during childhood and
adulthood combined40. This accounts for a large proportion of the reported twin heritabilities
estimated at 20-62%7,9–12. Reported trauma shows genetic correlations with psychiatric
disorders, current mental state, personality traits, lifestyle factors, and sociodemographic
traits21,39,40. However, these studies did not analytically explain the extent to which the h2
SNP of
reported traumas reflects genetic correlations with these complex traits. Identifying specific
traits that explain a large proportion of h2