Hostname: page-component-848d4c4894-2pzkn Total loading time: 0 Render date: 2024-05-31T00:15:41.583Z Has data issue: false hasContentIssue false

Genetic contributions to bipolar disorder: current status and future directions

Published online by Cambridge University Press:  21 April 2021

Kevin S. O'Connell*
Affiliation:
Division of Mental Health and Addiction, NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, 0407Oslo, Norway
Brandon J. Coombes
Affiliation:
Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
*
Author for correspondence: Kevin S. O'Connell, E-mail: kevin.oconnell@medisin.uio.no
Rights & Permissions [Opens in a new window]

Abstract

Bipolar disorder (BD) is a highly heritable mental disorder and is estimated to affect about 50 million people worldwide. Our understanding of the genetic etiology of BD has greatly increased in recent years with advances in technology and methodology as well as the adoption of international consortiums and large population-based biobanks. It is clear that BD is also highly heterogeneous and polygenic and shows substantial genetic overlap with other psychiatric disorders. Genetic studies of BD suggest that the number of associated loci is expected to substantially increase in larger future studies and with it, improved genetic prediction of the disorder. Still, a number of challenges remain to fully characterize the genetic architecture of BD. First among these is the need to incorporate ancestrally-diverse samples to move research away from a Eurocentric bias that has the potential to exacerbate health disparities already seen in BD. Furthermore, incorporation of population biobanks, registry data, and electronic health records will be required to increase the sample size necessary for continued genetic discovery, while increased deep phenotyping is necessary to elucidate subtypes within BD. Lastly, the role of rare variation in BD remains to be determined. Meeting these challenges will enable improved identification of causal variants for the disorder and also allow for equitable future clinical applications of both genetic risk prediction and therapeutic interventions.

Type
Invited Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re- use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press

Definition of illness

Affective disorders are classified along a spectrum from unipolar depression to bipolar disorder (BD) type II and type I (Carvalho, Firth, & Vieta, Reference Carvalho, Firth and Vieta2020; Grande, Berk, Birmaher, & Vieta, Reference Grande, Berk, Birmaher and Vieta2016). The presence of recurring manic or hypomanic episodes alternating with euthymia or depressive episodes distinguishes BD from other affective disorders (American Psychiatric Association, 2013; World Health Organization et al., 1992). BD type I (BDI) is characterized by alternating manic and depressive episodes (Fig. 1). Psychotic symptoms also occur in a majority of these patients which may lead to compromised functioning and hospitalization. The Diagnostic and Statistical Manual of Mental Disorder, Fifth Edition (DSM-5) also allows for individuals impaired by manic episodes without depression to still be diagnosed with BDI (American Psychiatric Association, 2013). In comparison, a diagnosis of BD type II (BDII) is based on the occurrence of at least one depressive and one hypomanic episode during the lifetime, but no manic episodes (Fig. 1). A diagnosis of BD not otherwise specified or BD unspecified may be given when a patient has bipolar symptoms that do not fit within these major subtype categories. The DSM-5 also includes specifiers which define the clinical features of episodes and the course of the disorder, namely, anxious distress, mixed features, rapid cycling, melancholic features, atypical features, psychotic features (mood-congruent and mood-incongruent), catatonia, peripartum onset, and seasonal pattern (American Psychiatric Association, 2013). In addition, the DSM-5 includes schizoaffective BD as a distinct diagnosis wherein individuals suffer from psychotic symptoms as well as episodes of mania or depression (American Psychiatric Association, 2013).

Fig. 1. Polarity of symptoms for bipolar disorder subtypes. Bipolar disorder type I is characterized by at least one manic episode. Bipolar disorder type II is characterized by at least one depressive and one hypomanic episode during the lifetime, but no manic episodes. Major depressive disorder does not include episodes of hypomania or mania.

Epidemiology

BD is projected to affect about 50 million people worldwide (GBD 2016 Disease and Injury Incidence and Prevalence Collaborators, 2017). The BD subtypes each have an estimated lifetime prevalence of approximately 1% (Merikangas et al., Reference Merikangas, Akiskal, Angst, Greenberg, Hirschfeld, Petukhova and Kessler2007, Reference Merikangas, Jin, He, Kessler, Lee, Sampson, Viana and Zarkov2011) although large ranges in lifetime prevalence have been reported (BDI: 0.1–1.7%, BDII: 0.1–3.0%) (Angst, Reference Angst1998; Merikangas et al., Reference Merikangas, Akiskal, Angst, Greenberg, Hirschfeld, Petukhova and Kessler2007, Reference Merikangas, Jin, He, Kessler, Lee, Sampson, Viana and Zarkov2011). Most studies report no gender differences in the prevalence of BD; however, women may be at increased risk for BDII, rapid cycling, and mixed episodes (Diflorio & Jones, Reference Diflorio and Jones2010; Nivoli et al., Reference Nivoli, Pacchiarotti, Rosa, Popovic, Murru, Valenti and Colom2011). The mean age of onset of BD is at approximately 20 years. An earlier age of onset is associated with poorer prognosis, increased comorbidity, onset beginning with depression, and more severe depressive episodes, as well as longer treatment delays (Joslyn, Hawes, Hunt, & Mitchell, Reference Joslyn, Hawes, Hunt and Mitchell2016). Additionally, initial depressive episodes may lead to a misdiagnosis of major depressive disorder until the onset of manic or hypomanic episodes necessary to confirm BD (Zimmerman, Ruggero, Chelminski, & Young, Reference Zimmerman, Ruggero, Chelminski and Young2008).

BD is often comorbid with other psychiatric (Eser, Kacar, Kilciksiz, Yalçinay-Inan, & Ongur, Reference Eser, Kacar, Kilciksiz, Yalçinay-Inan and Ongur2018; Frías, Baltasar, & Birmaher, Reference Frías, Baltasar and Birmaher2016; Salloum & Brown, Reference Salloum and Brown2017) and non-psychiatric disorders (Bortolato, Berk, Maes, McIntyre, & Carvalho, Reference Bortolato, Berk, Maes, McIntyre and Carvalho2016; Correll et al., Reference Correll, Solmi, Veronese, Bortolato, Rosson, Santonastaso and Stubbs2017; Vancampfort et al., Reference Vancampfort, Correll, Galling, Probst, De Hert, Ward and Stubbs2016). It is estimated that >90% of BD patients have at least one lifetime comorbid disorder, and >70% present with three or more comorbid disorders during their lifetime (Merikangas et al., Reference Merikangas, Akiskal, Angst, Greenberg, Hirschfeld, Petukhova and Kessler2007). Such ubiquitous comorbidity within BD suggests the disturbance of multiple systems and pathways, and the presence of comorbidities is associated with increased premature mortality in BD when compared to the general population (Kessing, Vradi, McIntyre, & Andersen, Reference Kessing, Vradi, McIntyre and Andersen2015; Roshanaei-Moghaddam & Katon, Reference Roshanaei-Moghaddam and Katon2009).

Classical genetic epidemiology

Family studies

Genetic factors for BD were first investigated using twin, family, and adoption studies. These studies established that family history of BD is an important clinical predictor of The onset of mood disorders in a patient and that the risk of mood disorder decreases as the genetic distance from the proband increases (Craddock & Sklar, Reference Craddock and Sklar2013; Merikangas & Yu, Reference Merikangas and Yu2002). A large family-based Swedish study showed the risk of BD was as much as 7.9, 3.3, and 1.6 times higher for first-, second-, and third-degree relatives of BD probands, respectively, than those without a proband in their family (Song et al., Reference Song, Bergen, Kuja-Halkola, Larsson, Landén and Lichtenstein2015). In the largest family study to date in the Swedish cohort with over 2 million individuals, the transmission of BD was found to be statistically homogenous across family type (intact family, families without fathers, and adoptive families) (Kendler, Ohlsson, Sundquist, & Sundquist, Reference Kendler, Ohlsson, Sundquist and Sundquist2020). This family-based study also estimated the heritability of BD, which is a measure of the proportion of variation in a given trait attributed to genetic variation, to be 44% (95% CI 36–48%). Estimates of heritability from twin studies, which compare the concordance of disease between monozygotic and dizygotic twins, were between 60% and 90% (Craddock & Sklar, Reference Craddock and Sklar2013; Merikangas & Yu, Reference Merikangas and Yu2002; Smoller & Finn, Reference Smoller and Finn2003). Furthermore, by comparing estimates from twin studies, the heritability of BD is among the highest of all other psychiatric and behavioral disorders (Bienvenu, Davydow, & Kendler, Reference Bienvenu, Davydow and Kendler2011).

It has also been well-established that familial risk of BD correlates with increased familial risk of other psychiatric disorders such as schizophrenia, depression, anxiety, drug abuse, attention-deficit hyperactivity disorder (ADHD), personality disorders, and autism spectrum disorder (ASD) (Craddock & Sklar, Reference Craddock and Sklar2013; Kendler et al., Reference Kendler, Ohlsson, Sundquist and Sundquist2020; Song et al., Reference Song, Bergen, Kuja-Halkola, Larsson, Landén and Lichtenstein2015). Schizophrenia, ASD, and depression have the strongest genetic correlations with BD as identified through family studies.

Cohort and population studies

Sample sizes of studies have rapidly increased as genetic studies of BD have moved from family-based designs to cohort and population-based designs. With these, computationally-efficient methods, such as linkage disequilibrium score regression (LDSC), have been developed to estimate both heritability and genetic correlation captured by the single nucleotide polymorphisms (SNPs) which are common locations in the genome where variation occurs between individuals and are measured on a genotyping array (Bulik-Sullivan et al., Reference Bulik-Sullivan, Loh, Finucane, Ripke, Yang and Neale2015a).

Using this technique, the largest study of BD estimated an SNP-based heritability (h 2SNP), which is a measure of the proportion of variation in a given trait attributed to the genetic variation captured by a genotyping array, of 18.6% (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). Figure 2 compares the latest estimates of the twin-based heritability and h 2SNP for BD to a range of psychiatric, behavioral, and neurological disorders (Bienvenu et al., Reference Bienvenu, Davydow and Kendler2011; Browne, Gair, Scharf, & Grice, Reference Browne, Gair, Scharf and Grice2014; Cross-Disorder Group of the Psychiatric Genomics Consortium, 2019; Demontis et al., Reference Demontis, Rajagopal, Thorgeirsson, Als, Grove, Leppälä and Børglum2019a; Faraone & Larsson, Reference Faraone and Larsson2019; Gatz et al., Reference Gatz, Reynolds, Fratiglioni, Johansson, Mortimer, Berg and Pedersen2006; Jansen et al., Reference Jansen, Savage, Watanabe, Bryois, Williams, Steinberg and Posthuma2019; Purves et al., Reference Purves, Coleman, Meier, Rayner, Davis, Cheesman and Eley2020; Walters et al., Reference Walters, Polimanti, Johnson, McClintick, Adams, Adkins and Agrawal2018; Zilhão et al., Reference Zilhão, Olthof, Smit, Cath, Ligthart, Mathews and Dolan2017). BD has the greatest twin-based heritability estimate and, similar to other traits, also has a substantial proportion not captured by common variations.

Fig. 2. Estimated heritability of psychiatric, behavioral, and neurological disorders. Mean twin-based and SNP-based (on liability scale) heritability for different psychiatric (BIP, bipolar disorder; SCZ, schizophrenia; ADHD, attention-deficit/hyperactivity disorder; MD, major depression; ANX, generalized anxiety disorder), behavioral (AN, anorexia nervosa; AUD, alcohol use disorder; CUD, cannabis use disorder), or neurological (ASD, autism spectrum disorder; AD, Alzheimer's disorder; OCD, obsessive-compulsive disorder; TS, Tourette's syndrome) disorders. Error bars are shown for SNP-based estimates from LDSC.

New methodologies have also allowed the genetic correlation between different traits to be estimated using the summary statistics from overlapping sets of common SNPs in different samples. A recent cross-disorder analysis of eight different psychiatric and neurological disorders found that BD is most strongly genetically correlated with schizophrenia (r g = 0.70) but also shares substantial genetic overlap with major depression (r g = 0.36), obsessive-compulsive disorder (OCD; r g = 0.31), anorexia nervosa (r g = 0.21), ADHD (r g = 0.14), and ASD (r g = 0.14) (Cross-Disorder Group of the Psychiatric Genomics Consortium, 2019). Furthermore, the high comorbidity of substance use in BD has been linked to substantial genetic correlation with substance use: cigarette use (r g = 0.1), cannabis use (r g = 0.27), and alcohol use disorder (r g = 0.30) (Jang et al., Reference Jang, Saunders, Liu, Jiang, Liu and Vrieze2020; Kranzler et al., Reference Kranzler, Zhou, Kember, Vickers Smith, Justice, Damrauer and Gelernter2019).

Molecular genetic epidemiology

Common variants

Genome-wide association studies (GWASs) have been the most successful strategy for identifying specific genetic variants associated with BD. Unlike linkage studies, a GWAS can be performed on a set of unrelated cases and controls by testing for the association of genetic variants across the genome with a trait. The first GWAS of BD in 2007 included 2000 cases and 3000 controls and only identified one independent association signal at p < 5 × 10–7 (Wellcome Trust Case Control Consortium, 2007). This association was below the now established p < 5 × 10–8 threshold for genome-wide significance and further, this association was not supported in an expanded reference group analysis nor in an independent replication. Since then, dozens of additional GWASs of case–control cohorts, consisting of samples from mostly European ancestries, have been published (Table 1). Until the sample size of the GWASs increased to over 10 K, these GWASs identified very few genome-wide significant loci. The most recent GWAS was based on a meta-analysis of 52 case–control cohorts and five large population-based cohorts and included over 40 K cases and 350 K controls identified 64 independent loci across the genome associated with BD (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). Thirty-three of these loci were novel for BD, including the region containing the major histocompatibility complex which is also strongly associated with schizophrenia (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020; Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2014). Furthermore, additional novel loci identified in this GWAS are also reported as risk loci for schizophrenia, depression, childhood-onset psychiatric disorders, and problematic alcohol use, highlighting shared underlying genetic architecture between BD and these other psychiatric disorders (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). Still though, the SNPs in this large study only explain 15–18% of the variance in the trait (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020).

Table 1. Summary of bipolar disorder GWAS

Furthermore, as shown in Fig. 3, univariate causal mixture modeling suggests that we can expect to see substantial increases in identified genome-wide significant loci and consequently in the proportion of h 2SNP explained by these variants as GWAS sample sizes increase (Holland et al., Reference Holland, Frei, Desikan, Fan, Shadrin, Smeland and Dale2020). This is particularly relevant for BD where GWAS studies have now reached the ‘inflection’ point where the significant associations begin to accumulate with smaller increases in sample size (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). As such, international collaborations in large-scale GWAS remain imperative for the continued identification of common variants underlying BD etiology, and the plan of the PGC Bipolar Working Group to further increase GWAS sample sizes is encouraging (Sullivan et al., Reference Sullivan, Agrawal, Bulik, Andreassen, Børglum, Breen and O'Donovan2018).

Fig. 3. Statistical power calculations for current and future GWAS. The variance explained by genome-wide significant variants (y-axis) is calculated for increasing GWAS sample sizes (x-axis) using the univariate causal mixture model (Holland et al., Reference Holland, Frei, Desikan, Fan, Shadrin, Smeland and Dale2020). The legend describes the estimated GWAS sample sizes (SE) needed to capture 50% of the genetic variance (horizontal dashed line) associated with each trait. Stars indicate the sample sizes of currently available GWAS, and circles indicate the estimated sample sizes needed to capture 50% of the genetic variance for each trait. Traits include attention-deficit/hyperactivity disorder (ADHD) (Demontis et al., Reference Demontis, Walters, Martin, Mattheisen, Als, Agerbo and Neale2019b), autism spectrum disorder (ASD) (Grove et al., Reference Grove, Ripke, Als, Mattheisen, Walters, Won and Børglum2019), bipolar disorder (BD) (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020), depression (MDD) (Howard et al., Reference Howard, Adams, Clarke, Hafferty, Gibson, Shirali and McIntosh2019), and schizophrenia (SCZ) (Pardiñas et al., Reference Pardiñas, Holmans, Pocklington, Escott-Price, Ripke, Carrera and Walters2018). Height is included as a somatic control (no genetic correlation exists between height and bipolar disorder) (Yengo et al., Reference Yengo, Sidorenko, Kemper, Zheng, Wood and Weedon2018). s.e., standard error.

Genetic overlap

In addition to genetic correlation (Bulik-Sullivan et al., Reference Bulik-Sullivan, Finucane, Anttila, Gusev, Day, Loh and Neale2015b) (described above), the most common approach for assessing genetic overlap at the genome-wide level is polygenic risk score (PRS) analysis (International Schizophrenia Consortium et al., Reference Purcell, Wray, Stone, Visscher, O'Donovan and Sklar2009). The PRS for a given trait is typically a weighted sum of genetic variants where the variants used and their weights are defined by effects measured by previous GWASs of the trait. The genetic liability for BD has been used to predict a number of other psychiatric disorders as well as creativity, educational attainment (Mistry, Harrison, Smith, Escott-Price, & Zammit, Reference Mistry, Harrison, Smith, Escott-Price and Zammit2018), addiction (Reginsson et al., Reference Reginsson, Ingason, Euesden, Bjornsdottir, Olafsson, Sigurdsson and Stefansson2018), as well as psychopathology (Mistry, Escott-Price, Florio, Smith, & Zammit, Reference Mistry, Escott-Price, Florio, Smith and Zammit2019a), cognitive functioning (Mistry, Escott-Price, Florio, Smith, & Zammit, Reference Mistry, Escott-Price, Florio, Smith and Zammit2019b), progression of unipolar to bipolar depression, and depression onset (Musliner et al., Reference Musliner, Mortensen, McGrath, Suppli, Hougaard and Bybjerg-Grauholm2019, Reference Musliner, Krebs, Albiñana, Vilhjalmsson, Agerbo, Zandi and Østergaard2020).

PRSs for BD and other traits have also been used to explain common comorbidities within BD. Suicide attempts by people with BD have been associated with higher genetic liability for depression (Mullins et al., Reference Mullins, Bigdeli, Børglum, Coleman, Demontis, Mehta and Lewis2019) as well as an interaction between trauma and bipolar genetic liability (Wilcox et al., Reference Wilcox, Fullerton, Glowinski, Benke, Kamali, Hulvershorn and Nurnberger2017). Previous childhood ADHD diagnosis in those with BD was associated with higher genetic liability for ADHD (Grigoroiu-Serbanescu et al., Reference Grigoroiu-Serbanescu, Giaroli, Thygesen, Shenyan, Bigdeli, Bass and McQuillin2020; Wilcox et al., Reference Wilcox, Fullerton, Glowinski, Benke, Kamali, Hulvershorn and Nurnberger2017).

In addition to PRS analysis, cross-disorder GWAS meta-analyses have also been performed for BD and ADHD (Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2018; van Hulzen et al., Reference van Hulzen, Scholz, Franke, Ripke, Klein, McQuillin and Reif2017), SCZ (Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2018), and MDD (Coleman, Gaspar, Bryois, & Breen, Reference Coleman, Gaspar, Bryois and Breen2020), identifying two, 114, and 73 genome-wide significant loci associated with these phenotype pairs, respectively. Moreover, numerous genes mapped to BD risk loci are also linked to schizophrenia, ASD, and OCD (O'Connell, McGregor, Lochner, Emsley, & Warnich, Reference O'Connell, McGregor, Lochner, Emsley and Warnich2018) further highlighting common genetic architecture across psychiatric disorders.

The conditional/conjunctional false discovery rate statistical tool has also been used to identify BD risk loci as well as shared risk loci between BD and a number of phenotypes. This method leverages the power of two GWAS to boost discovery by re-adjusting the GWAS test statistics in a primary phenotype and allows for the discovery of loci significantly associated with two phenotypes simultaneously (Andreassen, Thompson, & Dale, Reference Andreassen, Thompson and Dale2014; Smeland et al., Reference Smeland, Frei, Shadrin, O'Connell, Fan, Bahrami and Andreassen2020b). Utilizing this method, shared genetic loci have been identified between BD and ADHD (n = 5) (O'Connell et al., Reference O'Connell, Shadrin, Bahrami, Smeland, Bettella, Frei and Andreassen2019), schizophrenia (n = 14) (Andreassen et al., Reference Andreassen, Thompson, Schork, Ripke, Mattingsdal, Kelsoe and Dale2013), Alzheimer's disease (n = 2) (Drange et al., Reference Drange, Smeland, Shadrin, Finseth, Witoelar, Frei and Andreassen2019), intelligence (n = 12) (Smeland et al., Reference Smeland, Bahrami, Frei, Shadrin, O'Connell, Savage and Andreassen2020a), body mass index (n = 17) (Bahrami et al., Reference Bahrami, Steen, Shadrin, O'Connell, Frei, Bettella and Andreassen2020), and lifespan (n = 8) (Muntané et al., Reference Muntané, Farré, Bosch, Martorell, Navarro and Vilella2021). This method is agnostic to the effect directions of genetic variants and so shared loci were identified between BD and Alzheimer's disease, intelligence, body mass index, and lifespan despite observed null and non-significant genetic correlations with these phenotypes.

Most recently (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020), the genetic relationship between BD and 10 clinically and epidemiologically associated traits (daytime sleepiness, morningness, sleep duration, insomnia, mood instability, educational attainment, problematic alcohol use, drinks per week, smoking initiation, and cigarettes per day) were assessed using the MiXeR tool (Frei et al., Reference Frei, Holland, Smeland, Shadrin, Fan, Maeland and Dale2019), to identify trait-specific and shared genetic components, and Mendelian randomization (Zhu et al., Reference Zhu, Zheng, Zhang, Wu, Trzaskowski, Maier and Yang2018), to establish ‘causal’ relationships. Extensive genetic overlap was identified between all traits and BD, most notably that >90% of the genetic variants estimated to influence BD were also estimated to influence educational attainment. Moreover, bidirectional relationships were identified between BD and sleep duration, mood instability, educational attainment, and problematic alcohol use, while BD was identified as ‘causal’ for morningness and drinks per week and smoking initiation was ‘causal’ for BD (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020).

Rare variants

In addition to genetic interactions, the difference in heritability could also be explained by rare variants in the genome which are often unmeasured and thus not included in GWASs. While the cost of whole-exome sequencing (WES) and whole-genome sequencing (WGS) has decreased, these technologies are still substantially more expensive than common genotyping arrays. As a result, WGS/WES studies of BD have been limited to small studies consisting mostly of large pedigrees to potentially enrich the sample with causal rare variants and increase power (Forstner et al., Reference Forstner, Fischer, Schenk, Strohmaier, Maaser-Hecker, Reinbold and Cichon2020; Goes et al., Reference Goes, Pirooznia, Parla, Kramer, Ghiban, Mavruk and Potash2016, Reference Goes, Pirooznia, Tehan, Zandi, McGrath, Wolyniec and Pulver2019; Maaser et al., Reference Maaser, Forstner, Strohmaier, Hecker, Ludwig and Sivalingam2018; Sul et al., Reference Sul, Service, Huang, Ramensky, Hwang, Teshiba and Freimer2020; Toma et al., Reference Toma, Shaw, Allcock, Heath, Pierce, Mitchell and Fullerton2018). While these studies have found evidence of higher rare deleterious burden in cases (Sul et al., Reference Sul, Service, Huang, Ramensky, Hwang, Teshiba and Freimer2020), higher disruptive variant burden in early-onset cases (Toma et al., Reference Toma, Shaw, Allcock, Heath, Pierce, Mitchell and Fullerton2018), evidence of rare variant segregation in pedigrees (Forstner et al., Reference Forstner, Fischer, Schenk, Strohmaier, Maaser-Hecker, Reinbold and Cichon2020; Goes et al., Reference Goes, Pirooznia, Parla, Kramer, Ghiban, Mavruk and Potash2016; Maaser et al., Reference Maaser, Forstner, Strohmaier, Hecker, Ludwig and Sivalingam2018), and evidence of de novo variation (Goes et al., Reference Goes, Pirooznia, Tehan, Zandi, McGrath, Wolyniec and Pulver2019), much larger sample sizes will be required to definitively identify rare variants conferring risk for BD.

Copy number variants

Copy number variants (CNVs) refer to regions of the genome where a duplication (three or more copies are present) or deletion (only one copy remains) has occurred such that more or less than the expected two copies in the diploid human genome are present. Carriers of certain CNVs are known to be at considerably elevated risk for developing neurodevelopmental (e.g. ASDs) and mental disorders (e.g. schizophrenia) (Kirov, Rees, & Walters, Reference Kirov, Rees and Walters2015) as well as somatic conditions (e.g. diabetes and hypertension) (Crawford et al., Reference Crawford, Bracher-Smith, Owen, Kendall, Rees, Pardiñas and Kirov2019). The frequency of CNVs in BD is less than that observed for neurodevelopmental disorders or schizophrenia (Kirov, Reference Kirov2015), and correspondingly their role in the disorder appears less with only one CNV robustly associated with BD to date. A 650 kb duplication at 16p11.2 was first described as a de novo CNV for BD (Kirov, Reference Kirov2015; Malhotra et al., Reference Malhotra, McCarthy, Michaelson, Vacic, Burdick, Yoon and Sebat2011) and this association was replicated in a larger genome-wide analysis (Green et al., Reference Green, Rees, Walters, Smith, Forty, Grozeva and Kirov2016). This CNV is also implicated in schizophrenia, autism, and intellectual disability (Kirov, Reference Kirov2015). Two additional CNVs, at 1q21.1 and 3q29, are also implicated in BD; however, these associations fail to pass the genome-wide significance threshold (Green et al., Reference Green, Rees, Walters, Smith, Forty, Grozeva and Kirov2016). Interestingly, these two CNVs are also associated with schizophrenia (Kirov, Reference Kirov2015). One further study identified enrichment of genic CNVs in schizoaffective BD, but not between BD cases and controls or other BD subtypes (Charney et al., Reference Charney, Stahl, Green, Chen, Moran, Chambert and Sklar2019).

These findings highlight that the genetic overlap between BD and schizophrenia extends beyond common variation, but suggests a difference in underlying mechanisms. One possible explanation for the smaller role of CNVs in BD is that patients with BD exhibit less cognitive deficits than patients with schizophrenia who can exhibit substantial cognitive deficits, since the same CNVs which are implicated in schizophrenia are also known to cause cognitive problems (Kirov, Reference Kirov2015).

Genetic interactions

Other than increasing the sample size of GWAS, the difference between observed twin-based and h 2SNP (Fig. 2) may also be explained by unaccounted for moderated genetic effects such as interactions between genes and the environment (G×E) or gene–gene interactions (epistatic effects). The role of G×E in BD remains an under-researched area, however, but some interactions have been identified (Aas et al., Reference Aas, Haukvik, Djurovic, Tesli, Athanasiu, Bjella and Melle2014, Reference Aas, Bellivier, Bettella, Henry, Gard, Kahn and Etain2020; Hosang, Fisher, Cohen-Woods, McGuffin, & Farmer, Reference Hosang, Fisher, Cohen-Woods, McGuffin and Farmer2017; Oliveira et al., Reference Oliveira, Kazma, Le Floch, Bennabi, Hamdani, Bengoufa and Tamouza2016; Winham et al., Reference Winham, Cuellar-Barboza, Oliveros, McElroy, Crow, Colby and Biernacka2014). Although these studies highlight the potential role of G×E in the etiology of BD, the lack of replication studies and small sample sizes suggest that they should be interpreted with caution. As with G×E, studies of epistasis in BD are in their infancy and lack replication (Judy et al., Reference Judy, Seifuddin, Pirooznia, Mahon, Jancic, Goes and Zandi2013). As the ability of GWAS to identify risk variants with small effects increases, further study of how implicated genes interact with environmental or other genetic factors to modulate the risk of BD are required.

Clinical implications

Pharmacogenomics

Lithium, anti-epileptic drug mood stabilizers (such as valproate/divalproex, lamotrigine, and carbamazepine), antipsychotics, and antidepressants are commonly prescribed treatments for BD. However, response to these medications can widely vary between individuals, and some patients may cycle through different medications before they find an effective treatment with minimal side effects. Pharmacogenomic studies aim to use genetics to predict treatment response. A particular challenge to pharmacogenomics in BD has been the measurement of treatment response which can be limited by the length of follow-up, adherence to medication, and confounding due to the multi-drug treatment strategy common to the illness. Consequently, a systematic rating system with a high inter-rater reliability, the Alda score, was developed to quantify the clinical improvement of BD during treatment while also accounting for potential confounders of treatment response (Nunes, Trappenberg, & Alda, Reference Nunes, Trappenberg and Alda2020). However, obtaining large samples with reliable measures has limited the statistical power to discover clinically-informative genetic variants associated with treatment response. Furthermore, heterogeneity between study designs and the samples included have yielded limited replication of any findings. While not yet replicable, promising pharmacogenomic findings for BD were summarized in a recent review (Gordovez & McMahon, Reference Gordovez and McMahon2020). Most of the previous pharmacogenomic studies have been focused on either lithium treatment response or HLA haplotypes predicting serious adverse reactions related to carbamazepine, phenytoin, and lamotrigine. A recent study tested for genetic association with treatment response to anti-epileptic drug mood stabilizers, an alternative to lithium, and identified two SNP-level associations in THSD7A and SLC35F3 as well as two gene-level associations with ABCC1 and DISP1 (Ho et al., Reference Ho, Coombes, Nguyen, Liu, McElroy, Singh and Biernacka2020).

With the exception of genetic predictors of adverse reactions to medication, no large genetic effects on treatment response have been identified. However, current pharmacogenomic testing has already been shown to be useful by providing clinicians support in reaching effective and well-tolerated treatments of BD (Ielmini et al., Reference Ielmini, Poloni, Caselli, Espadaler, Tuson, Grecchi and Callegari2018). Additionally, as the sample size of pharmacogenomic studies increases, PRSs derived from these studies could further enable a precision medicine approach to BD treatment. In addition to pharmacogenomic PRSs, PRSs derived from large case–control studies could also improve the genetic prediction of treatment response. For example, increased genetic liability for depression and schizophrenia was associated with worse response to lithium (Amare et al., Reference Amare, Schubert, Hou, Clark, Papiol, Cearns and Baune2020; International Consortium on Lithium Genetics (ConLi+Gen) et al., Reference Amare, Schubert, Hou, Clark, Papiol and Baune2018). These PRSs could be explaining some of the clinical heterogeneity in the sample as discussed below and thus improve the identification of certain BD clinical profiles that respond best to lithium.

Finally, there is potential application of repurposing drugs and focusing on different drug targets based on recent genetic findings. For example, calcium channel blockers (CBBs), which have been widely used to treat hypertension and other cardiovascular conditions, were also once considered as a treatment in psychiatry (Harrison, Tunbridge, Dolphin, & Hall, Reference Harrison, Tunbridge, Dolphin and Hall2020). However, because CACNA1C has now been implicated as one of the strongest associations with BD (Gordovez & McMahon, Reference Gordovez and McMahon2020), there is renewed interest in CBBs as a treatment for the disorder (Cipriani et al., Reference Cipriani, Saunders, Attenburrow, Stefaniak, Panchal, Stockton and Harrison2016).

Risk prediction

In addition to therapeutic intervention, PRSs may also provide clinical utility to inform disease screening (Torkamani, Wineinger, & Topol, Reference Torkamani, Wineinger and Topol2018). While the PRS derived from the latest GWAS of BD only explains about 4.75% of the phenotypic variance, the latest PRS could still be useful for risk stratification (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). Compared to individuals with average genetic risk for BD, individuals in the top decile risk had an odds ratio of 3.62 (95% CI 1.7–7.9) of being a case. An important caveat to note about PRSs, however, is that prediction performance is worse when applied to ancestries not included in the training GWAS (Martin et al., Reference Martin, Kanai, Kamatani, Okada, Neale and Daly2019). For instance, the current BD PRS, estimated using individuals with European ancestries, explains only around 2% and 1% of the phenotypic variance in individuals with East Asian or admixed African American ancestry, respectively (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). Encouragingly though, the trans-ethnic prediction accuracy of the PRS has improved as the sample size has increased. Furthermore, the PRS prediction accuracy will also improve as new non-European ancestries are included in future training GWASs.

Clinical heterogeneity

PRSs can also help dissect the high clinical heterogeneity (i.e. bipolar type, psychosis, rapid cycling) present in the disorder (Coombes et al., n.d.). For example, higher genetic liability for schizophrenia is associated with bipolar type I (Charney et al., Reference Charney, Ruderfer, Stahl, Moran, Chambert, Belliveau and Sklar2017). This finding could be driven by the increased prevalence of psychosis among those with BDI as multiple studies have shown that higher genetic risk of schizophrenia is associated with psychosis in BD, particularly during mania (Allardyce et al., Reference Allardyce, Leonenko, Hamshere, Pardiñas, Forty, Knott and Escott-Price2018; Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2018; Charney et al., Reference Charney, Stahl, Green, Chen, Moran, Chambert and Sklar2019; Coombes et al., Reference Coombes, Markota, Mann, Colby, Stahl, Talati and Biernacka2020; Markota et al., Reference Markota, Coombes, Larrabee, McElroy, Bond, Veldic and Biernacka2018). Other studies of bipolar subtypes have shown positive associations between BDII and insomnia PRS, rapid cycling and ADHD PRS, as well as early age-of-onset of BD and PRSs for risk-taking and anhedonia (Coombes et al., n.d.; Lewis et al., Reference Lewis, Richards, Karlsson, Leonenko, Jones, Jones and Di Florio2019). While no individual PRS is able to explain a large amount of variation among bipolar subtypes, these findings give insight into the genetic contributions to clinical heterogeneity and could help classify the disorder more accurately as well as identify the risk of suicide, psychosis, and other adverse outcomes in patients with BD.

Future directions

Significant advances in our understanding of the genetic architecture of BD have been made, from initial linkage and family studies to current large consortia-driven genome-wide studies. Moreover, integration of these genetic discoveries with other -omic and imaging data will be key to comprehending the role of genetic variation in the etiology of BD. However, distinct shortcomings and limitations to genetic discovery highlight key areas to be prioritized in future studies.

Diverse phenotype ascertainment

Identification of novel loci for BD, and other polygenic complex phenotypes, requires increasing sample sizes (Fig. 3), which remains a challenging and costly task (Lu, Campeau, & Lee, Reference Lu, Campeau and Lee2014). The majority of samples included in the PGC-BD were clinically ascertained, with the inclusion of external biobank samples only in the most recent discovery GWAS (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). Numerous efforts have been made to combine electronic health record and registry data with genetic data to facilitate large population-based studies, such as the Electronic Medical Records and Genomics network (https://emerge-network.org/), the UK Biobank (https://www.ukbiobank.ac.uk/), All of Us (https://allofus.nih.gov/), the Million Veterans Program (https://www.research.va.gov/mvp/), and iPsych (https://ipsych.dk/en/). Furthermore, GWAS summary statistics of self-reported phenotypes for thousands to millions of individuals may be obtained through collaboration with the personal genetics company 23andMe, Inc. (https://research.23andme.com/research-innovation-collaborations/). The data generated by such population studies and 23andMe provide a means by which to drastically increase sample size without the costs associated with clinical ascertainment. This approach was shown to be successful for depression, where PGC cohorts were meta-analyzed with data from the UK Biobank and summary statistics from 23andMe, increasing the number of identified associated risk loci from 44 (Wray et al., Reference Wray, Ripke, Mattheisen, Trzaskowski, Byrne and Abdellaoui2018) to 102 (Howard et al., Reference Howard, Adams, Clarke, Hafferty, Gibson, Shirali and McIntosh2019). However, a limitation to this use of ‘minimal phenotyping’ data is that the loci identified, especially when based on self-report data, were non-specific for depression highlighting potential differences in genetic architecture when compared to clinically ascertained depression (Cai et al., Reference Cai, Revez, Adams, Andlauer, Breen, Byrne and Flint2020). In line with this, the h 2SNP estimates of the biobank samples included in the latest PGC BD GWAS are less than that observed for clinically ascertained samples which may reflect more heterogeneous clinical presentations or less severe illness (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020).

Data generated from ‘minimal phenotyping’ are likely to include other psychopathological features which may underlie self-reported BD such as personality disorders or mild temperamental traits, thereby increasing heterogeneity in the sample and leading to the possibility of non-specific or false-positive results. However, true self-reported BD may reflect the non-hospitalized, non-psychotic part of the BD spectrum, more typical of BDII, which is under-represented in the current PGC BD sample. Moreover, expanding genetic studies to include the full spectrum of BD in population-based non-clinical samples increases the potential for novel discoveries with important implications for clinical management and further research, and is therefore of high interest to both clinicians and the pharmaceutical industry.

Thus, while adopting the ‘minimal phenotyping’ approach for BD will allow GWAS to reach sample sizes not currently feasible by clinical ascertainment and will likely identify numerous novel risk loci, similar post-hoc analyses as that performed for depression (Cai et al., Reference Cai, Revez, Adams, Andlauer, Breen, Byrne and Flint2020), will be required to determine the specificity of identified loci to BD.

Increased deep phenotyping

The high levels of heterogeneity amongst patients with BD, including disorder type, features of episodes, and the course of the disorder, contribute to the difficulty in identifying underlying genetic risk factors. BDI (h 2SNP = 25%) is shown to be more heritable than BDII (h 2SNP = 11%), and the genetic correlation (r g = 0.89) between these types suggests that they are closely related, yet distinct, phenotypes (Stahl et al., Reference Stahl, Breen, Forstner, McQuillin, Ripke and Trubetskoy2019). In support of this, the most recent PGC GWAS for BD identified novel and distinct loci specifically associated with BDI or BDII, which were not identified when all bipolar cases were analyzed together (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). Genetic studies of the features and course of BD have predominantly employed a PRS approach, as outlined above, and GWAS data for these subtypes is lacking due to small sample sizes [data from the PGC indicate that none of these subtypes include more than 10 K samples (Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2018)]. Thus, larger deeply phenotyped samples are required in order to conduct a thorough investigation of the genetic architecture of these subtypes within BD. Doing so would aid subtype-specific discoveries, and may inform on nosology, diagnostic practices, and drug development for BD.

In addition, the potential inclusion of ‘minimal phenotyping’ data, as described above, further emphasizes the need for increased deep phenotyping. Results generated from deep phenotyped samples will serve as standards against which to compare the specificity of results generated from the inclusion of ‘minimal phenotyping’.

Increased ancestral-diversity

The majority of individuals included in GWASs for any trait have overwhelmingly been of European descent and the lack of diversity is even more pronounced in genetic studies of psychiatric disorders (Martin et al., Reference Martin, Kanai, Kamatani, Okada, Neale and Daly2019; Peterson et al., Reference Peterson, Kuchenbaecker, Walters, Chen, Popejoy, Periyasamy and Duncan2019; Sirugo, Williams, & Tishkoff, Reference Sirugo, Williams and Tishkoff2019). In BD, the largest GWAS includes only individuals from European ancestries (Mullins et al., Reference Mullins, Forstner, O'Connell, Coombes, Coleman, Qiao and Andreassen2020). This ‘missing diversity’ can greatly hinder our understanding of the etiology of BD. For example, the inclusion of non-European ancestries could substantially improve fine-mapping of disease-associated loci (Peterson et al., Reference Peterson, Kuchenbaecker, Walters, Chen, Popejoy, Periyasamy and Duncan2019). Furthermore, the current Eurocentric approach has the potential to exacerbate health disparities already seen in BD (Akinhanmi et al., Reference Akinhanmi, Biernacka, Strakowski, McElroy, Balls Berry, Merikangas and Frye2018) by limiting the therapeutic advances gained by pharmacogenomics and improved genetic risk predictions to those of European descent (Duncan et al., Reference Duncan, Shen, Gelaye, Meijsen, Ressler, Feldman and Domingue2019; Martin et al., Reference Martin, Kanai, Kamatani, Okada, Neale and Daly2019; Sirugo et al., Reference Sirugo, Williams and Tishkoff2019). Future inclusion of diverse samples will come with new ethical, technological, and methodological challenges (Peterson et al., Reference Peterson, Kuchenbaecker, Walters, Chen, Popejoy, Periyasamy and Duncan2019). Some of these considerations include choosing ancestry-specific genotyping platforms to improve genomic coverage, increasing sample sizes of diverse reference panels to improve imputation accuracy, and improving statistical methods to control for population stratification and estimate ancestry-specific PRSs. Thus, the PGC Bipolar Working Group has committed to expanding the future GWAS to include non-European ancestries.

Larger sequencing efforts

As mentioned above, sequencing efforts in BD are currently in their infancy (Forstner et al., Reference Forstner, Fischer, Schenk, Strohmaier, Maaser-Hecker, Reinbold and Cichon2020; Goes et al., Reference Goes, Pirooznia, Parla, Kramer, Ghiban, Mavruk and Potash2016; Maaser et al., Reference Maaser, Forstner, Strohmaier, Hecker, Ludwig and Sivalingam2018; Sul et al., Reference Sul, Service, Huang, Ramensky, Hwang, Teshiba and Freimer2020; Toma et al., Reference Toma, Shaw, Allcock, Heath, Pierce, Mitchell and Fullerton2018). Although studies provide evidence that rare variants might contribute to the etiology of BD, weak statistical power due to small sample sizes remains an issue. The Bipolar Sequencing Consortium (BSC) was established to facilitate combining existing exome and WGS studies of BD (http://metamoodics.org/bsc/consortium/), and includes approximately 4500 BD cases and 9000 controls, as well as 1200 affected relatives from 250 families. Moreover, a collaboration between the Dalio Initiative in BD (https://www.daliophilanthropies.org/initiatives/mental-health-and-wellness/), the Stanley Centre (https://www.broadinstitute.org/stanley), and iPSYCH (https://ipsych.dk/en/) aims to generate WES data from approximately 7000 BD cases and 10 000 matched controls. However, it is estimated that as many as 25 000 cases might be necessary in order to identify significant rare variant associations with BD (Zuk et al., Reference Zuk, Schaffner, Samocha, Do, Hechter, Kathiresan and Lander2014), confirmed by recent analyses in schizophrenia (Singh et al., Reference Singh, Poterba, Curtis, Akil, Al Eissa, Barchas and Daly2020), and so continued expansion of these, or similar, efforts will be crucial to determine the role of rare variation in BD.

Conclusion

Our knowledge of the genetic etiology of BD has rapidly accelerated in recent years with advances in technology and methodology as well as the adoption of international consortiums and large population-based biobanks. It is now clear that BD is highly heritable but also highly heterogeneous and polygenic with substantial genetic overlap with other psychiatric disorders. Encouragingly, genetic studies of BD have reached an ‘inflection point’ (Fig. 3). Thus, the number of associated loci is expected to substantially increase in larger future studies and with it, improved genetic prediction of the disorder. Incorporation of ancestrally-diverse samples in these studies will enable improved identification of causal variants for the disorder and also allow for equitable future clinical applications of both genetic risk prediction and therapeutic interventions.

Acknowledgements

We would like to thank the research participants and members of the Bipolar Disorder Working Group of the Psychiatric Genomics Consortium, and other studies reported in this review, for making this research possible.

Financial support

We acknowledge the support from the Research Council of Norway (229129, 213837, 223273), the South-East Norway Regional Health Authority (2017-112), and the PGC US Norway Collaboration (RCN# 248980).

Conflict of interest

None.

References

Aas, M., Bellivier, F., Bettella, F., Henry, C., Gard, S., Kahn, J.-P.Etain, B. (2020). Childhood maltreatment and polygenic risk in bipolar disorders. Bipolar Disorders, 22(2), 174181. https://doi.org/10.1111/bdi.12851.CrossRefGoogle ScholarPubMed
Aas, M., Haukvik, U. K., Djurovic, S., Tesli, M., Athanasiu, L., Bjella, T.Melle, I. (2014). Interplay between childhood trauma and BDNF val66met variants on blood BDNF mRNA levels and on hippocampus subfields volumes in schizophrenia spectrum and bipolar disorders. Journal of Psychiatric Research, 59, 1421. https://doi.org/10.1016/j.jpsychires.2014.08.011.CrossRefGoogle ScholarPubMed
Akinhanmi, M. O., Biernacka, J. M., Strakowski, S. M., McElroy, S. L., Balls Berry, J. E., Merikangas, K. R.Frye, M. A. (2018). Racial disparities in bipolar disorder treatment and research: A call to action. Bipolar Disorders, 20(6), 506514. https://doi.org/10.1111/bdi.12638.CrossRefGoogle ScholarPubMed
Allardyce, J., Leonenko, G., Hamshere, M., Pardiñas, A. F., Forty, L., Knott, S.Escott-Price, V. (2018). Association between schizophrenia-related polygenic liability and the occurrence and level of mood-incongruent psychotic symptoms in bipolar disorder. JAMA Psychiatry, 75(1), 2835. https://doi.org/10.1001/jamapsychiatry.2017.3485.CrossRefGoogle ScholarPubMed
International Consortium on Lithium Genetics (ConLi + Gen), Amare, A. T., Schubert, K. O., Hou, L., Clark, S. R., Papiol, S.Baune, B. T. (2018). Association of polygenic score for schizophrenia and HLA antigen and inflammation genes with response to lithium in bipolar affective disorder: A genome-wide association study. JAMA Psychiatry, 75(1), 6574. https://doi.org/10.1001/jamapsychiatry.2017.3433.Google ScholarPubMed
Amare, A. T., Schubert, K. O., Hou, L., Clark, S. R., Papiol, S., Cearns, M.Baune, B. T. (2020). Association of polygenic score for major depression with response to lithium in patients with bipolar disorder. Molecular Psychiatry. https://doi.org/10.1038/s41380-020-0689-5.Google ScholarPubMed
American Psychiatric Association. (2013). Diagnostic and statistical manual of mental disorders (5th ed.). Washington, DC 20024, USA: American Psychiatric Association Publishing. https://books.google.com/books/about/Diagnostic_and_Statistical_Manual_of_Men.html?hl=&id=-JivBAAAQBAJ.Google Scholar
Andreassen, O. A., Thompson, W. K., & Dale, A. M. (2014). Boosting the power of schizophrenia genetics by leveraging new statistical tools. Schizophrenia Bulletin, 40(1), 1317. https://doi.org/10.1093/schbul/sbt168.CrossRefGoogle ScholarPubMed
Andreassen, O. A., Thompson, W. K., Schork, A. J., Ripke, S., Mattingsdal, M., Kelsoe, J. R.Dale, A. M. (2013). Improved detection of common variants associated with schizophrenia and bipolar disorder using pleiotropy-informed conditional false discovery rate. PLoS Genetics, 9(4), e1003455. https://doi.org/10.1371/journal.pgen.1003455.CrossRefGoogle ScholarPubMed
Angst, J. (1998). The emerging epidemiology of hypomania and bipolar II disorder. Journal of Affective Disorders, 50(2–3), 143151. https://doi.org/10.1016/s0165-0327(98)00142-6.CrossRefGoogle ScholarPubMed
Bahrami, S., Steen, N. E., Shadrin, A., O'Connell, K., Frei, O., Bettella, F.Andreassen, O. A. (2020). Shared genetic loci between body mass index and major psychiatric disorders: A genome-wide association study. JAMA Psychiatry, 77(5), 503512. https://doi.org/10.1001/jamapsychiatry.2019.4188.CrossRefGoogle ScholarPubMed
Baum, A. E., Akula, N., Cabanero, M., Cardona, I., Corona, W., Klemens, B.McMahon, F. J. (2008). A genome-wide association study implicates diacylglycerol kinase eta (DGKH) and several other genes in the etiology of bipolar disorder. Molecular Psychiatry, 13(2), 197207. https://doi.org/10.1038/sj.mp.4002012.CrossRefGoogle ScholarPubMed
Bergen, S. E., O'Dushlaine, C. T., Ripke, S., Lee, P. H., Ruderfer, D. M., Akterin, S.Sullivan, P. F. (2012). Genome-wide association study in a Swedish population yields support for greater CNV and MHC involvement in schizophrenia compared with bipolar disorder. Molecular Psychiatry 17(9), 880886. https://doi.org/10.1038/mp.2012.73.CrossRefGoogle Scholar
Bienvenu, O. J., Davydow, D. S., & Kendler, K. S. (2011). Psychiatric ‘diseases’ versus behavioral disorders and degree of genetic influence. Psychological Medicine, 41(1), 3340. https://doi.org/10.1017/S003329171000084X.CrossRefGoogle ScholarPubMed
Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium. (2018). Genomic dissection of bipolar disorder and schizophrenia, including 28 subphenotypes. Cell, 173(7), 17051715.e16. https://doi.org/10.1016/j.cell.2018.05.046.CrossRefGoogle Scholar
Bortolato, B., Berk, M., Maes, M., McIntyre, R. S., & Carvalho, A. F. (2016). Fibromyalgia and bipolar disorder: Emerging epidemiological associations and shared pathophysiology. Current Molecular Medicine, 16(2), 119136. https://doi.org/10.2174/1566524016666160126144027.CrossRefGoogle ScholarPubMed
Browne, H. A., Gair, S. L., Scharf, J. M., & Grice, D. E. (2014). Genetics of obsessive-compulsive disorder and related disorders. The Psychiatric Clinics of North America, 37(3), 319335. https://doi.org/10.1016/j.psc.2014.06.002.CrossRefGoogle ScholarPubMed
Bulik-Sullivan, B., Finucane, H. K., Anttila, V., Gusev, A., Day, F. R., Loh, P.-R.Neale, B. M. (2015b). An atlas of genetic correlations across human diseases and traits. Nature Genetics, 47(11), 12361241. https://doi.org/10.1038/ng.3406.CrossRefGoogle Scholar
Bulik-Sullivan, B. K, & , Loh, P.-R., Finucane, H. K., Ripke, S., Yang, J., & Schizophrenia Working Group of the Psychiatric Genomics ConsortiumNeale, B. M., & (2015a). LD score regression distinguishes confounding from polygenicity in genome-wide association studies. Nature Genetics 47(3), 291295. https://doi.org/10.1038/ng.3211.CrossRefGoogle Scholar
Cai, N., Revez, J. A., Adams, M. J., Andlauer, T. F. M., Breen, G., Byrne, E. M.Flint, J. (2020). Minimal phenotyping yields genome-wide association signals of low specificity for major depression. Nature Genetics, 52(4), 437447. https://doi.org/10.1038/s41588-020-0594-5.CrossRefGoogle ScholarPubMed
Carvalho, A. F., Firth, J., & Vieta, E. (2020). Bipolar disorder. The New England Journal of Medicine, 383(1), 5866. https://doi.org/10.1056/NEJMra1906193.CrossRefGoogle ScholarPubMed
Charney, A. W., Ruderfer, D. M., Stahl, E. A., Moran, J. L., Chambert, K., Belliveau, R. A.Sklar, P. (2017). Evidence for genetic heterogeneity between clinical subtypes of bipolar disorder. Translational Psychiatry, 7(1), e993. https://doi.org/10.1038/tp.2016.242.CrossRefGoogle ScholarPubMed
Charney, A. W., Stahl, E. A., Green, E. K., Chen, C.-Y., Moran, J. L., Chambert, K.Sklar, P. (2019). Contribution of rare copy number variants to bipolar disorder risk is limited to schizoaffective cases. Biological Psychiatry, 86(2), 110119. https://doi.org/10.1016/j.biopsych.2018.12.009.CrossRefGoogle ScholarPubMed
Chen, D. T., Jiang, X., Akula, N., Shugart, Y. Y., Wendland, J. R., Steele, C. J. M.Strauss, J. (2013). Genome-wide association study meta-analysis of European and Asian-ancestry samples identifies three novel loci associated with bipolar disorder. Molecular Psychiatry, 18(2), 195205. https://doi.org/10.1038/mp.2011.157.CrossRefGoogle ScholarPubMed
Cichon, S., Mühleisen, T. W., Degenhardt, F. A., Mattheisen, M., Miró, X., Strohmaier, J.Nöthen, M. M. (2011). Genome-wide association study identifies genetic variation in neurocan as a susceptibility factor for bipolar disorder. American Journal of Human Genetics, 88(3), 372381. https://doi.org/10.1016/j.ajhg.2011.01.017.CrossRefGoogle ScholarPubMed
Cipriani, A., Saunders, K., Attenburrow, M.-J., Stefaniak, J., Panchal, P., Stockton, S.Harrison, P. J. (2016). A systematic review of calcium channel antagonists in bipolar disorder and some considerations for their future development. Molecular Psychiatry, 21(10), 13241332. https://doi.org/10.1038/mp.2016.86.CrossRefGoogle ScholarPubMed
Coleman, J. R. I., Gaspar, H. A., Bryois, J., Bipolar Disorder Working Group of the Psychiatric Genomics Consortium, Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium, & Breen, G. (2020). The genetics of the mood disorder Spectrum: Genome-wide association analyses of more than 185 000 cases and 439 000 controls. Biological Psychiatry, 88(2), 169184. https://doi.org/10.1016/j.biopsych.2019.10.015.CrossRefGoogle ScholarPubMed
Coombes, B., Markota, M., Mann, J., Colby, C., Stahl, E., Talati, A.Biernacka, J. M. (2020). Dissecting clinical heterogeneity of bipolar disorder using multiple polygenic risk scores. medRxiv, 2020.03.31.20044800. https://doi.org/10.1101/2020.03.31.20044800.CrossRefGoogle Scholar
Correll, C. U., Solmi, M., Veronese, N., Bortolato, B., Rosson, S., Santonastaso, P.Stubbs, B. (2017). Prevalence, incidence and mortality from cardiovascular disease in patients with pooled and specific severe mental illness: A large-scale meta-analysis of 3 211 768 patients and 113 383 368 controls. World Psychiatry: Official Journal of the World Psychiatric Association, 16(2), 163180. https://doi.org/10.1002/wps.20420.CrossRefGoogle ScholarPubMed
Craddock, N., & Sklar, P. (2013). Genetics of bipolar disorder. The Lancet, 381(9878), 16541662. https://doi.org/10.1016/S0140-6736(13)60855-7.CrossRefGoogle ScholarPubMed
Crawford, K., Bracher-Smith, M., Owen, D., Kendall, K. M., Rees, E., Pardiñas, A. F.Kirov, G. (2019). Medical consequences of pathogenic CNVs in adults: Analysis of the UK Biobank. Journal of Medical Genetics, 56(3), 131138. https://doi.org/10.1136/jmedgenet-2018-105477.CrossRefGoogle ScholarPubMed
Cross-Disorder Group of the Psychiatric Genomics Consortium. (2019). Genomic relationships, novel loci, and pleiotropic mechanisms across eight psychiatric disorders. Cell, 179(7), 14691482.e11. https://doi.org/10.1016/j.cell.2019.11.020.CrossRefGoogle Scholar
Demontis, D., Rajagopal, V. M., Thorgeirsson, T. E., Als, T. D., Grove, J., Leppälä, K.Børglum, A. D. (2019a). Genome-wide association study implicates CHRNA2 in cannabis use disorder. Nature Neuroscience, 22(7), 10661074. https://doi.org/10.1038/s41593-019-0416-1.CrossRefGoogle Scholar
Demontis, D., Walters, R. K., Martin, J., Mattheisen, M., Als, T. D., Agerbo, E.Neale, B. M. (2019b). Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nature Genetics, 51(1), 6375. https://doi.org/10.1038/s41588-018-0269-7.CrossRefGoogle Scholar
Diflorio, A., & Jones, I. (2010). Is sex important? Gender differences in bipolar disorder. International Review of Psychiatry, 22(5), 437452. https://doi.org/10.3109/09540261.2010.514601.CrossRefGoogle ScholarPubMed
Djurovic, S., Gustafsson, O., Mattingsdal, M., Athanasiu, L., Bjella, T., Tesli, M.Andreassen, O. A. (2010). A genome-wide association study of bipolar disorder in Norwegian individuals, followed by replication in Icelandic sample. Journal of Affective Disorders, 126(1–2), 312316. https://doi.org/10.1016/j.jad.2010.04.007.CrossRefGoogle ScholarPubMed
Drange, O. K., Smeland, O. B., Shadrin, A. A., Finseth, P. I., Witoelar, A., Frei, O.Andreassen, O. A. (2019). Genetic overlap between Alzheimer's disease and bipolar disorder implicates the MARK2 and VAC14 genes. Frontiers in Neuroscience 13, 220. https://doi.org/10.3389/fnins.2019.00220.CrossRefGoogle ScholarPubMed
Duncan, L., Shen, H., Gelaye, B., Meijsen, J., Ressler, K., Feldman, M.Domingue, B. (2019). Analysis of polygenic risk score usage and performance in diverse human populations. Nature Communications 10(1), 3328. https://doi.org/10.1038/s41467-019-11112-0.CrossRefGoogle ScholarPubMed
Eser, H. Y., Kacar, A. S., Kilciksiz, C. M., Yalçinay-Inan, M., & Ongur, D. (2018). Prevalence and associated features of anxiety disorder comorbidity in bipolar disorder: A meta-analysis and meta-regression study. Frontiers in Psychiatry, 9, 229. https://doi.org/10.3389/fpsyt.2018.00229.CrossRefGoogle Scholar
Faraone, S. V., & Larsson, H. (2019). Genetics of attention deficit hyperactivity disorder. Molecular Psychiatry, 24(4), 562575. https://doi.org/10.1038/s41380-018-0070-0.CrossRefGoogle ScholarPubMed
Ferreira, M. A. R., O'Donovan, M. C., Meng, Y. A., Jones, I. R., Ruderfer, D. M., Jones, L. … Wellcome Trust Case Control Consortium. (2008). Collaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorder. Nature Genetics, 40(9), 10561058. https://doi.org/10.1038/ng.209.CrossRefGoogle ScholarPubMed
Fiorica, P. N., & Wheeler, H. E. (2019). Transcriptome association studies of neuropsychiatric traits in African Americans implicate PRMT7 in schizophrenia. PeerJ 7, e7778. https://doi.org/10.7717/peerj.7778.CrossRefGoogle Scholar
Forstner, A. J., Fischer, S. B., Schenk, L. M., Strohmaier, J., Maaser-Hecker, A., Reinbold, C. S.Cichon, S. (2020). Whole-exome sequencing of 81 individuals from 27 multiply affected bipolar disorder families. Translational Psychiatry, 10(1), 57. https://doi.org/10.1038/s41398-020-0732-y.CrossRefGoogle ScholarPubMed
Frei, O., Holland, D., Smeland, O. B., Shadrin, A. A., Fan, C. C., Maeland, S.Dale, A. M. (2019). Bivariate causal mixture model quantifies polygenic overlap between complex traits beyond genetic correlation. Nature Communications, 10(1), 2417. https://doi.org/10.1038/s41467-019-10310-0.CrossRefGoogle ScholarPubMed
Frías, Á., Baltasar, I., & Birmaher, B. (2016). Comorbidity between bipolar disorder and borderline personality disorder: Prevalence, explanatory theories, and clinical impact. Journal of Affective Disorders, 202, 210219. https://doi.org/10.1016/j.jad.2016.05.048.CrossRefGoogle ScholarPubMed
Gatz, M., Reynolds, C. A., Fratiglioni, L., Johansson, B., Mortimer, J. A., Berg, S.Pedersen, N. L. (2006). Role of genes and environments for explaining Alzheimer disease. Archives of General Psychiatry, 63(2), 168174. https://doi.org/10.1001/archpsyc.63.2.168.CrossRefGoogle ScholarPubMed
GBD 2016 Disease and Injury Incidence and Prevalence Collaborators. (2017). Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: A systematic analysis for the Global Burden of Disease Study 2016. The Lancet, 390(10100), 12111259. https://doi.org/10.1016/S0140-6736(17)32154-2.CrossRefGoogle Scholar
Goes, F. S., Pirooznia, M., Parla, J. S., Kramer, M., Ghiban, E., Mavruk, S.Potash, J. B. (2016). Exome sequencing of familial bipolar disorder. JAMA Psychiatry, 73(6), 590597. https://doi.org/10.1001/jamapsychiatry.2016.0251.CrossRefGoogle ScholarPubMed
Goes, F. S., Pirooznia, M., Tehan, M., Zandi, P. P., McGrath, J., Wolyniec, P.Pulver, A. E. (2019). De novo variation in bipolar disorder. Molecular Psychiatry, 387, 1561. https://doi.org/10.1038/s41380-019-0611-1.Google Scholar
Gordovez, F. J. A., & McMahon, F. J. (2020). The genetics of bipolar disorder. Molecular Psychiatry, 25(3), 544559. https://doi.org/10.1038/s41380-019-0634-7.CrossRefGoogle ScholarPubMed
Grande, I., Berk, M., Birmaher, B., & Vieta, E. (2016). Bipolar disorder. The Lancet, 387(10027), 15611572. https://doi.org/10.1016/S0140-6736(15)00241-X.CrossRefGoogle ScholarPubMed
Green, E. K., Rees, E., Walters, J. T. R., Smith, K.-G., Forty, L., Grozeva, D.Kirov, G. (2016). Copy number variation in bipolar disorder. Molecular Psychiatry, 21(1), 8993. https://doi.org/10.1038/mp.2014.174.CrossRefGoogle ScholarPubMed
Grigoroiu-Serbanescu, M., Giaroli, G., Thygesen, J. H., Shenyan, O., Bigdeli, T. B., Bass, N. J.McQuillin, A. (2020). Predictive power of the ADHD GWAS 2019 polygenic risk scores in independent samples of bipolar patients with childhood ADHD. Journal of Affective Disorders, 265, 651659. https://doi.org/10.1016/j.jad.2019.11.109.CrossRefGoogle ScholarPubMed
Grove, J., Ripke, S., Als, T. D., Mattheisen, M., Walters, R. K., Won, H.Børglum, A. D. (2019). Identification of common genetic risk variants for autism spectrum disorder. Nature Genetics, 51(3), 431444. https://doi.org/10.1038/s41588-019-0344-8.CrossRefGoogle ScholarPubMed
Harrison, P. J., Tunbridge, E. M., Dolphin, A. C., & Hall, J. (2020). Voltage-gated calcium channel blockers for psychiatric disorders: Genomic reappraisal. The British Journal of Psychiatry: The Journal of Mental Science, 216(5), 250253. https://doi.org/10.1192/bjp.2019.157.CrossRefGoogle ScholarPubMed
Hattori, E., Toyota, T., Ishitsuka, Y., Iwayama, Y., Yamada, K., Ujike, H.Yoshikawa, T. (2009). Preliminary genome-wide association study of bipolar disorder in the Japanese population. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics: The Official Publication of the International Society of Psychiatric Genetics, 150B(8), 11101117. https://doi.org/10.1002/ajmg.b.30941.CrossRefGoogle ScholarPubMed
Ho, A. M.-C., Coombes, B. J., Nguyen, T. T. L., Liu, D., McElroy, S. L., Singh, B.Biernacka, J. M. (2020). Mood-stabilizing antiepileptic treatment response in bipolar disorder: A genome-wide association study. Clinical Pharmacology and Therapeutics, 108(6), 12331242. https://doi.org/10.1002/cpt.1982.CrossRefGoogle ScholarPubMed
Holland, D., Frei, O., Desikan, R., Fan, C.-C., Shadrin, A. A., Smeland, O. B.Dale, A. M. (2020). Beyond SNP heritability: Polygenicity and discoverability of phenotypes estimated with a univariate Gaussian mixture model. PLoS Genetics, 16(5), e1008612. https://doi.org/10.1371/journal.pgen.1008612.CrossRefGoogle ScholarPubMed
Hosang, G. M., Fisher, H. L., Cohen-Woods, S., McGuffin, P., & Farmer, A. E. (2017). Stressful life events and catechol-O-methyl-transferase (COMT) gene in bipolar disorder. Depression and Anxiety, 34(5), 419426. https://doi.org/10.1002/da.22606.CrossRefGoogle ScholarPubMed
Hou, L., Bergen, S. E., Akula, N., Song, J., Hultman, C. M., Landén, M.McMahon, F. J. (2016). Genome-wide association study of 40000 individuals identifies two novel loci associated with bipolar disorder. Human Molecular Genetics, 25(15), 33833394. https://doi.org/10.1093/hmg/ddw181.CrossRefGoogle ScholarPubMed
Howard, D. M., Adams, M. J., Clarke, T.-K., Hafferty, J. D., Gibson, J., Shirali, M.McIntosh, A. M. (2019). Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nature Neuroscience, 22(3), 343352. https://doi.org/10.1038/s41593-018-0326-7.CrossRefGoogle ScholarPubMed
Ielmini, M., Poloni, N., Caselli, I., Espadaler, J., Tuson, M., Grecchi, A., & Callegari, C. (2018). The utility of pharmacogenetic testing to support the treatment of bipolar disorder. Pharmacogenomics and Personalized Medicine, 11, 3542. https://doi.org/10.2147/PGPM.S160967.CrossRefGoogle ScholarPubMed
Ikeda, M., Takahashi, A., Kamatani, Y., Okahisa, Y., Kunugi, H., Mori, N.Iwata, N. (2018). A genome-wide association study identifies two novel susceptibility loci and trans population polygenicity associated with bipolar disorder. Molecular Psychiatry, 23(3), 639647. https://doi.org/10.1038/mp.2016.259.CrossRefGoogle ScholarPubMed
Jang, S.-K., Saunders, G., Liu, M., Jiang, Y., Liu, D. J., & Vrieze, S., & 23andMe Research Team. (2020). Genetic correlation, pleiotropy, and causal associations between substance use and psychiatric disorder. Psychological Medicine, 111. https://doi.org/10.1017/s003329172000272x.CrossRefGoogle ScholarPubMed
Jansen, I. E., Savage, J. E., Watanabe, K., Bryois, J., Williams, D. M., Steinberg, S.Posthuma, D. (2019). Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer's disease risk. Nature Genetics, 51(3), 404413. https://doi.org/10.1038/s41588-018-0311-9.CrossRefGoogle ScholarPubMed
Joslyn, C., Hawes, D. J., Hunt, C., & Mitchell, P. B. (2016). Is age of onset associated with severity, prognosis, and clinical features in bipolar disorder? A meta-analytic review. Bipolar Disorders, 18(5), 389403. https://doi.org/10.1111/bdi.12419.CrossRefGoogle ScholarPubMed
Judy, J. T., Seifuddin, F., Pirooznia, M., Mahon, P. B., Bipolar Genome Study Consortium, Jancic, D., Goes, F. S.Zandi, P. P. (2013). Converging evidence for epistasis between ANK3 and potassium channel gene KCNQ2 in bipolar disorder. Frontiers in Genetics, 4, 87. https://doi.org/10.3389/fgene.2013.00087.CrossRefGoogle ScholarPubMed
Kao, C.-F., Chen, H.-W., Chen, H.-C., Yang, J.-H., Huang, M.-C., Chiu, Y.-H.Kuo, P.-H. (2016). Identification of susceptible loci and enriched pathways for bipolar II disorder using genome-wide association studies. The International Journal of Neuropsychopharmacology/Official Scientific Journal of the Collegium Internationale Neuropsychopharmacologicum, 19(12). https://doi.org/10.1093/ijnp/pyw064.CrossRefGoogle ScholarPubMed
Kendler, K. S., Ohlsson, H., Sundquist, J., & Sundquist, K. (2020). An extended Swedish national adoption study of bipolar disorder illness and cross-generational familial association with schizophrenia and major depression. JAMA Psychiatry, 77(8), 814822. https://doi.org/10.1001/jamapsychiatry.2020.0223.CrossRefGoogle ScholarPubMed
Kerner, B., Lambert, C. G., & Muthén, B. O. (2011). Genome-wide association study in bipolar patients stratified by co-morbidity. PLoS ONE, 6(12), e28477. https://doi.org/10.1371/journal.pone.0028477.CrossRefGoogle ScholarPubMed
Kessing, L. V., Vradi, E., McIntyre, R. S., & Andersen, P. K. (2015). Causes of decreased life expectancy over the life span in bipolar disorder. Journal of Affective Disorders, 180, 142147. https://doi.org/10.1016/j.jad.2015.03.027.CrossRefGoogle ScholarPubMed
Kirov, G. (2015). CNVs in neuropsychiatric disorders. Human Molecular Genetics, 24(R1), R45R49. https://doi.org/10.1093/hmg/ddv253.CrossRefGoogle ScholarPubMed
Kirov, G., Rees, E., & Walters, J. (2015). What a psychiatrist needs to know about copy number variants. BJPsych Advances, 21(3), 157163. https://doi.org/10.1192/apt.bp.113.012039.CrossRefGoogle Scholar
Kranzler, H. R., Zhou, H., Kember, R. L., Vickers Smith, R., Justice, A. C., Damrauer, S.Gelernter, J. (2019). Genome-wide association study of alcohol consumption and use disorder in 274 424 individuals from multiple populations. Nature Communications, 10(1), 1499. https://doi.org/10.1038/s41467-019-09480-8.CrossRefGoogle ScholarPubMed
Kuo, P. H., Chuang, L. C., Liu, J. R., Liu, C. M., Huang, M. C., Lin, S. K.Lu, R. B. (2014). Identification of novel loci for bipolar I disorder in a multi-stage genome-wide association study. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 51, 5864. https://doi.org/10.1016/j.pnpbp.2014.01.003.CrossRefGoogle Scholar
Lee, H.-J., Woo, H. G., Greenwood, T. A., Kripke, D. F., & Kelsoe, J. R. (2013). A genome-wide association study of seasonal pattern mania identifies NF1A as a possible susceptibility gene for bipolar disorder. Journal of Affective Disorders, 145(2), 200207. https://doi.org/10.1016/j.jad.2012.07.032.CrossRefGoogle ScholarPubMed
Lewis, K. J. S., Richards, A., Karlsson, R., Leonenko, G., Jones, S. E., Jones, H. J.Di Florio, A. (2019). Comparison of genetic liability for sleep traits among individuals with bipolar disorder I or II and control participants. JAMA Psychiatry, 77(3), 303310. https://doi.org/10.1001/jamapsychiatry.2019.4079.CrossRefGoogle Scholar
Lu, J. T., Campeau, P. M., & Lee, B. H. (2014). Genotype–phenotype correlation – promiscuity in the era of next-generation sequencing. The New England Journal of Medicine, 371(7), 593596. https://doi.org/10.1056/NEJMp1400788.CrossRefGoogle ScholarPubMed
Maaser, A., Forstner, A. J., Strohmaier, J., Hecker, J., Ludwig, K. U., Sivalingam, S. … Bipolar Disorder Working Group of the Psychiatric Genomics Consortium. (2018). Exome sequencing in large, multiplex bipolar disorder families from Cuba. PLoS ONE, 13(10), e0205895. https://doi.org/10.1371/journal.pone.0205895.CrossRefGoogle ScholarPubMed
Malhotra, D., McCarthy, S., Michaelson, J. J., Vacic, V., Burdick, K. E., Yoon, S.Sebat, J. (2011). High frequencies of de novo CNVs in bipolar disorder and schizophrenia. Neuron, 72(6), 951963. https://doi.org/10.1016/j.neuron.2011.11.007.CrossRefGoogle Scholar
Markota, M., Coombes, B. J., Larrabee, B. R., McElroy, S. L., Bond, D. J., Veldic, M.Biernacka, J. M. (2018). Association of schizophrenia polygenic risk score with manic and depressive psychosis in bipolar disorder. Translational Psychiatry, 8(1), 188. https://doi.org/10.1038/s41398-018-0242-3.CrossRefGoogle ScholarPubMed
Martin, A. R., Kanai, M., Kamatani, Y., Okada, Y., Neale, B. M., & Daly, M. J. (2019). Clinical use of current polygenic risk scores may exacerbate health disparities. Nature Genetics, 51(4), 584591. https://doi.org/10.1038/s41588-019-0379-x.CrossRefGoogle ScholarPubMed
Merikangas, K. R., Akiskal, H. S., Angst, J., Greenberg, P. E., Hirschfeld, R. M. A., Petukhova, M., & Kessler, R. C. (2007). Lifetime and 12-month prevalence of bipolar spectrum disorder in the National Comorbidity Survey replication. Archives of General Psychiatry, 64(5), 543552. https://doi.org/10.1001/archpsyc.64.5.543.CrossRefGoogle ScholarPubMed
Merikangas, K. R., Jin, R., He, J.-P., Kessler, R. C., Lee, S., Sampson, N. A., Viana, M. C.Zarkov, Z. (2011). Prevalence and correlates of bipolar spectrum disorder in the world mental health survey initiative. Archives of General Psychiatry, 68(3), 241251. https://doi.org/10.1001/archgenpsychiatry.2011.12.CrossRefGoogle ScholarPubMed
Merikangas, K., & Yu, K. (2002). Genetic epidemiology of bipolar disorder. Clinical Neuroscience Research, 2(3–4), 127141. https://doi.org/10.1016/s1566-2772(02)00038-5.CrossRefGoogle Scholar
Mistry, S., Escott-Price, V., Florio, A. D., Smith, D. J., & Zammit, S. (2019a). Genetic risk for bipolar disorder and psychopathology from childhood to early adulthood. Journal of Affective Disorders, 246, 633639. https://doi.org/10.1016/j.jad.2018.12.091.CrossRefGoogle Scholar
Mistry, S., Escott-Price, V., Florio, A. D., Smith, D. J., & Zammit, S. (2019b). Investigating associations between genetic risk for bipolar disorder and cognitive functioning in childhood. Journal of Affective Disorders, 259, 112120. https://doi.org/10.1016/j.jad.2019.08.040.CrossRefGoogle Scholar
Mistry, S., Harrison, J. R., Smith, D. J., Escott-Price, V., & Zammit, S. (2018). The use of polygenic risk scores to identify phenotypes associated with genetic risk of bipolar disorder and depression: A systematic review. Journal of Affective Disorders, 234, 148155. https://doi.org/10.1016/j.jad.2018.02.005.CrossRefGoogle ScholarPubMed
Mühleisen, T. W., Leber, M., Schulze, T. G., Strohmaier, J., Degenhardt, F., Treutlein, J.Cichon, S. (2014). Genome-wide association study reveals two new risk loci for bipolar disorder. Nature Communications, 5, 3339. https://doi.org/10.1038/ncomms4339.CrossRefGoogle ScholarPubMed
Mullins, N., Bigdeli, T. B., Børglum, A. D., Coleman, J. R. I., Demontis, D., Mehta, D.Lewis, C. M. (2019). GWAS Of suicide attempt in psychiatric disorders and association with major depression polygenic risk scores. The American Journal of Psychiatry, 176(8), 651660. https://doi.org/10.1176/appi.ajp.2019.18080957.CrossRefGoogle ScholarPubMed
Mullins, N., Forstner, A. J., O'Connell, K. S., Coombes, B., Coleman, J. R. I., Qiao, Z.Andreassen, O. A. (2020). Genome-wide association study of over 40000 bipolar disorder cases provides novel biological insights. medRxiv, 2020.09.17.20187054. https://doi.org/10.1101/2020.09.17.20187054.CrossRefGoogle Scholar
Muntané, G., Farré, X., Bosch, E., Martorell, L., Navarro, A., & Vilella, E. (2021). The shared genetic architecture of schizophrenia, bipolar disorder and lifespan. Human Genetics, 140, 441455. https://doi.org/10.1007/s00439-020-02213-8.CrossRefGoogle ScholarPubMed
Musliner, K. L., Krebs, M. D., Albiñana, C., Vilhjalmsson, B., Agerbo, E., Zandi, P. P.Østergaard, S. D. (2020). Polygenic risk and progression to bipolar or psychotic disorders among individuals diagnosed with unipolar depression in early life. American Journal of Psychiatry, 177(10), 936943. https://doi.org/10.1176/appi.ajp.2020.19111195.CrossRefGoogle ScholarPubMed
Musliner, K. L., Mortensen, P. B., McGrath, J. J., Suppli, N. P., Hougaard, D. M., & Bybjerg-Grauholm, J. … Bipolar Disorder Working Group of the Psychiatric Genomics Consortium. (2019). Association of polygenic liabilities for major depression, bipolar disorder, and schizophrenia with risk for depression in the Danish population. JAMA Psychiatry, 76(5), 516525. https://doi.org/10.1001/jamapsychiatry.2018.4166.CrossRefGoogle ScholarPubMed
Nivoli, A. M. A., Pacchiarotti, I., Rosa, A. R., Popovic, D., Murru, A., Valenti, M.Colom, F. (2011). Gender differences in a cohort study of 604 bipolar patients: The role of predominant polarity. Journal of Affective Disorders, 133(3), 443449. https://doi.org/10.1016/j.jad.2011.04.055.CrossRefGoogle Scholar
Nunes, A., Trappenberg, T., Alda, M., & International Consortium on Lithium Genetics (ConLiGen). (2020). Asymmetrical reliability of the Alda score favours a dichotomous representation of lithium responsiveness. PLoS ONE, 15(1), e0225353. https://doi.org/10.1371/journal.pone.0225353.CrossRefGoogle ScholarPubMed
O'Connell, K. S., McGregor, N. W., Lochner, C., Emsley, R., & Warnich, L. (2018). The genetic architecture of schizophrenia, bipolar disorder, obsessive-compulsive disorder and autism spectrum disorder. Molecular and Cellular Neurosciences, 88, 300307. https://doi.org/10.1016/j.mcn.2018.02.010.CrossRefGoogle ScholarPubMed
O'Connell, K. S., Shadrin, A., Bahrami, S., Smeland, O. B., Bettella, F., Frei, O.Andreassen, O. A. (2019). Identification of genetic overlap and novel risk loci for attention-deficit/hyperactivity disorder and bipolar disorder. Molecular Psychiatry. https://doi.org/10.1038/s41380-019-0613-z.Google ScholarPubMed
Oliveira, J., Kazma, R., Le Floch, E., Bennabi, M., Hamdani, N., Bengoufa, D.Tamouza, R. (2016). Toxoplasma gondii exposure may modulate the influence of TLR2 genetic variation on bipolar disorder: A gene–environment interaction study. International Journal of Bipolar Disorders, 4(11). https://doi.org/10.1186/s40345-016-0052-6.CrossRefGoogle ScholarPubMed
Pardiñas, A. F., Holmans, P., Pocklington, A. J., Escott-Price, V., Ripke, S., Carrera, N.Walters, J. T. R. (2018). Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection. Nature Genetics, 50(3), 381389. https://doi.org/10.1038/s41588-018-0059-2.CrossRefGoogle ScholarPubMed
Peterson, R. E., Kuchenbaecker, K., Walters, R. K., Chen, C.-Y., Popejoy, A. B., Periyasamy, S.Duncan, L. E. (2019). Genome-wide association studies in ancestrally diverse populations: Opportunities, methods, pitfalls, and recommendations. Cell, 179(3), 589603. https://doi.org/10.1016/j.cell.2019.08.051.CrossRefGoogle Scholar
Psychiatric GWAS Consortium Bipolar Disorder Working Group. (2011). Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4. Nature Genetics, 43(10), 977983. https://doi.org/10.1038/ng.943.CrossRefGoogle Scholar
International Schizophrenia Consortium, Purcell, S. M., Wray, N. R., Stone, J. L., Visscher, P. M., O'Donovan, M. C.Sklar, P. (2009). Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature, 460(7256), 748752. https://doi.org/10.1038/nature08185.Google ScholarPubMed
Purves, K. L., Coleman, J. R. I., Meier, S. M., Rayner, C., Davis, K. A. S., Cheesman, R.Eley, T. C. (2020). A major role for common genetic variation in anxiety disorders. Molecular Psychiatry, 25, 32923303. https://doi.org/10.1038/s41380-019-0559-1.CrossRefGoogle Scholar
Reginsson, G. W., Ingason, A., Euesden, J., Bjornsdottir, G., Olafsson, S., Sigurdsson, E.Stefansson, K. (2018). Polygenic risk scores for schizophrenia and bipolar disorder associate with addiction. Addiction Biology, 23(1), 485492. https://doi.org/10.1111/adb.12496.CrossRefGoogle ScholarPubMed
Roshanaei-Moghaddam, B., & Katon, W. (2009). Premature mortality from general medical illnesses among persons with bipolar disorder: A review. Psychiatric Services, 60(2), 147156. https://doi.org/10.1176/ps.2009.60.2.147.CrossRefGoogle ScholarPubMed
Salloum, I. M., & Brown, E. S. (2017). Management of comorbid bipolar disorder and substance use disorders. The American Journal of Drug and Alcohol Abuse, 43(4), 366376. https://doi.org/10.1080/00952990.2017.1292279.CrossRefGoogle ScholarPubMed
Schizophrenia Working Group of the Psychiatric Genomics Consortium. (2014). Biological insights from 108 schizophrenia-associated genetic loci. Nature, 511(7510), 421427. https://doi.org/10.1038/nature13595.CrossRefGoogle Scholar
Scott, L. J., Muglia, P., Kong, X. Q., Guan, W., Flickinger, M., Upmanyu, R.Boehnke, M. (2009). Genome-wide association and meta-analysis of bipolar disorder in individuals of European ancestry. Proceedings of the National Academy of Sciences of the USA, 106(18), 75017506. https://doi.org/10.1073/pnas.0813386106.CrossRefGoogle ScholarPubMed
Singh, T., Poterba, T., Curtis, D., Akil, H., Al Eissa, M., Barchas, J. D.Daly, M. J. (2020). Exome sequencing identifies rare coding variants in 10 genes which confer substantial risk for schizophrenia. medRxiv, 2020.09.18.20192815. https://doi.org/10.1101/2020.09.18.20192815.CrossRefGoogle Scholar
Sirugo, G., Williams, S. M., & Tishkoff, S. A. (2019). The missing diversity in human genetic studies. Cell, 177(4), 1080. https://doi.org/10.1016/j.cell.2019.04.032.CrossRefGoogle ScholarPubMed
Sklar, P., Smoller, J. W., Fan, J., Ferreira, M. A. R., Perlis, R. H., Chambert, K.Purcell, S. M. (2008). Whole-genome association study of bipolar disorder. Molecular Psychiatry, 13(6), 558569. https://doi.org/10.1038/sj.mp.4002151.CrossRefGoogle ScholarPubMed
Smeland, O. B., Bahrami, S., Frei, O., Shadrin, A., O'Connell, K., Savage, J.Andreassen, O. A. (2020a). Genome-wide analysis reveals extensive genetic overlap between schizophrenia, bipolar disorder, and intelligence. Molecular Psychiatry, 25(4), 844853. https://doi.org/10.1038/s41380-018-0332-x.CrossRefGoogle Scholar
Smeland, O. B., Frei, O., Shadrin, A., O'Connell, K., Fan, C.-C., Bahrami, S.Andreassen, O. A. (2020b). Discovery of shared genomic loci using the conditional false discovery rate approach. Human Genetics, 139(1), 8594. https://doi.org/10.1007/s00439-019-02060-2.CrossRefGoogle Scholar
Smith, E. N., Bloss, C. S., Badner, J. A., Barrett, T., Belmonte, P. L., Berrettini, W.Kelsoe, J. R. (2009). Genome-wide association study of bipolar disorder in European American and African American individuals. Molecular Psychiatry, 14(8), 755763. https://doi.org/10.1038/mp.2009.43.CrossRefGoogle ScholarPubMed
Smith, E. N., Koller, D. L., Panganiban, C., Szelinger, S., Zhang, P., Badner, J. A.Kelsoe, J. R. (2011). Genome-wide association of bipolar disorder suggests an enrichment of replicable associations in regions near genes. PLoS Genetics, 7(6), e1002134. https://doi.org/10.1371/journal.pgen.1002134.CrossRefGoogle ScholarPubMed
Smoller, J. W., & Finn, C. T. (2003). Family, twin, and adoption studies of bipolar disorder. American Journal of Medical Genetics. Part C, Seminars in Medical Genetics, 123C(1), 4858. https://doi.org/10.1002/ajmg.c.20013.CrossRefGoogle ScholarPubMed
Song, J., Bergen, S. E., Kuja-Halkola, R., Larsson, H., Landén, M., & Lichtenstein, P. (2015). Bipolar disorder and its relation to major psychiatric disorders: A family-based study in the Swedish population. Bipolar Disorders, 17(2), 184193. https://doi.org/10.1111/bdi.12242.CrossRefGoogle ScholarPubMed
Stahl, E. A., Breen, G., Forstner, A. J., McQuillin, A., Ripke, S., Trubetskoy, V. … Bipolar Disorder Working Group of the Psychiatric Genomics Consortium. (2019). Genome-wide association study identifies 30 loci associated with bipolar disorder. Nature Genetics, 51(5), 793803. https://doi.org/10.1038/s41588-019-0397-8.CrossRefGoogle ScholarPubMed
Sul, J. H., Service, S. K., Huang, A. Y., Ramensky, V., Hwang, S.-G., Teshiba, T. M.Freimer, N. B. (2020). Contribution of common and rare variants to bipolar disorder susceptibility in extended pedigrees from population isolates. Translational Psychiatry, 10(1), 74. https://doi.org/10.1038/s41398-020-0758-1.CrossRefGoogle ScholarPubMed
Sullivan, P. F., Agrawal, A., Bulik, C. M., Andreassen, O. A., Børglum, A. D., Breen, G.O'Donovan, M. C. (2018). Psychiatric genomics: An update and an agenda. American Journal of Psychiatry, 175(1), 1527. https://doi.org/10.1176/appi.ajp.2017.17030283.CrossRefGoogle Scholar
Toma, C., Shaw, A. D., Allcock, R. J. N., Heath, A., Pierce, K. D., Mitchell, P. B.Fullerton, J. M. (2018). An examination of multiple classes of rare variants in extended families with bipolar disorder. Translational Psychiatry, 8(1), 65. https://doi.org/10.1038/s41398-018-0113-y.CrossRefGoogle ScholarPubMed
Torkamani, A., Wineinger, N. E., & Topol, E. J. (2018). The personal and clinical utility of polygenic risk scores. Nature Reviews. Genetics, 19(9), 581590. https://doi.org/10.1038/s41576-018-0018-x.CrossRefGoogle ScholarPubMed
Vancampfort, D., Correll, C. U., Galling, B., Probst, M., De Hert, M., Ward, P. B.Stubbs, B. (2016). Diabetes mellitus in people with schizophrenia, bipolar disorder and major depressive disorder: A systematic review and large scale meta-analysis. World Psychiatry: Official Journal of the World Psychiatric Association, 15(2), 166174. https://doi.org/10.1002/wps.20309.CrossRefGoogle ScholarPubMed
van Hulzen, K. J. E., Scholz, C. J., Franke, B., Ripke, S., Klein, M., McQuillin, A.Reif, A. (2017). Genetic overlap between attention-deficit/hyperactivity disorder and bipolar disorder: Evidence from genome-wide association study meta-analysis. Biological Psychiatry, 82(9), 634641. https://doi.org/10.1016/j.biopsych.2016.08.040.CrossRefGoogle ScholarPubMed
Walters, R. K., Polimanti, R., Johnson, E. C., McClintick, J. N., Adams, M. J., Adkins, A. E.Agrawal, A. (2018). Transancestral GWAS of alcohol dependence reveals common genetic underpinnings with psychiatric disorders. Nature Neuroscience, 21(12), 16561669. https://doi.org/10.1038/s41593-018-0275-1.CrossRefGoogle ScholarPubMed
Wellcome Trust Case Control Consortium. (2007). Genome-wide association study of 14 000 cases of seven common diseases and 3000 shared controls. Nature, 447(7145), 661678. https://doi.org/10.1038/nature05911.CrossRefGoogle Scholar
Wilcox, H. C., Fullerton, J. M., Glowinski, A. L., Benke, K., Kamali, M., Hulvershorn, L. A.Nurnberger, J. I. Jr. (2017). Traumatic stress interacts with bipolar disorder genetic risk to increase risk for suicide attempts. Journal of the American Academy of Child and Adolescent Psychiatry, 56(12), 10731080. https://doi.org/10.1016/j.jaac.2017.09.428.CrossRefGoogle ScholarPubMed
Winham, S. J., Cuellar-Barboza, A. B., Oliveros, A., McElroy, S. L., Crow, S., Colby, C.Biernacka, J. M. (2014). Genome-wide association study of bipolar disorder accounting for effect of body mass index identifies a new risk allele in TCF7L2. Molecular Psychiatry, 19(9), 10101016. https://doi.org/10.1038/mp.2013.159.CrossRefGoogle ScholarPubMed
World Health Organization, WHO Staff, & WHO. (1992). The ICD-10 classification of mental and behavioural disorders: Clinical descriptions and diagnostic guidelines. Geneva, Switzerland: World Health Organization. https://books.google.com/books/about/The_ICD_10_Classification_of_Mental_and.html?hl=&id=DFM0DgAAQBAJ.Google Scholar
Wray, N. R., Ripke, S., Mattheisen, M., Trzaskowski, M., Byrne, E. M., Abdellaoui, A. … Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium. (2018). Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nature Genetics, 50(5), 668681. https://doi.org/10.1038/s41588-018-0090-3.CrossRefGoogle ScholarPubMed
Xu, W., Cohen-Woods, S., Chen, Q., Noor, A., Knight, J., Hosang, G.Vincent, J. B. (2014). Genome-wide association study of bipolar disorder in Canadian and UK populations corroborates disease loci including SYNE1 and CSMD1. BMC Medical Genetics, 15, 2. https://doi.org/10.1186/1471-2350-15-2.CrossRefGoogle ScholarPubMed
Yengo, L., Sidorenko, J., Kemper, K. E., Zheng, Z., Wood, A. R., Weedon, M. N.GIANT Consortium. (2018). Meta-analysis of genome-wide association studies for height and body mass index in ~700 000 individuals of European ancestry. Human Molecular Genetics, 27(20), 36413649. https://doi.org/10.1093/hmg/ddy271.CrossRefGoogle Scholar
Yosifova, A., Mushiroda, T., Kubo, M., Takahashi, A., Kamatani, Y., Kamatani, N.Nakamura, Y. (2011). Genome-wide association study on bipolar disorder in the Bulgarian population. Genes, Brain, and Behavior, 10(7), 789797. https://doi.org/10.1111/j.1601-183X.2011.00721.x.CrossRefGoogle ScholarPubMed
Zhu, Z., Zheng, Z., Zhang, F., Wu, Y., Trzaskowski, M., Maier, R., … Yang, J. (2018). Causal associations between risk factors and common diseases inferred from GWAS summary data. Nature Communications, 9(1), 224. https://doi.org/10.1038/s41467-017-02317-2.CrossRefGoogle ScholarPubMed
Zilhão, N. R., Olthof, M. C., Smit, D. J. A., Cath, D. C., Ligthart, L., Mathews, C. A.Dolan, C. V. (2017). Heritability of tic disorders: A twin-family study. Psychological Medicine, 47(6), 10851096. https://doi.org/10.1017/S0033291716002981.CrossRefGoogle ScholarPubMed
Zimmerman, M., Ruggero, C. J., Chelminski, I., & Young, D. (2008). Is bipolar disorder overdiagnosed? The Journal of Clinical Psychiatry, 69(6), 935940. https://doi.org/10.4088/jcp.v69n0608.CrossRefGoogle ScholarPubMed
Zuk, O., Schaffner, S. F., Samocha, K., Do, R., Hechter, E., Kathiresan, S.Lander, E. S. (2014). Searching for missing heritability: Designing rare variant association studies. Proceedings of the National Academy of Sciences, 111(4), E455E464. https://doi.org/10.1073/pnas.1322563111.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Polarity of symptoms for bipolar disorder subtypes. Bipolar disorder type I is characterized by at least one manic episode. Bipolar disorder type II is characterized by at least one depressive and one hypomanic episode during the lifetime, but no manic episodes. Major depressive disorder does not include episodes of hypomania or mania.

Figure 1

Fig. 2. Estimated heritability of psychiatric, behavioral, and neurological disorders. Mean twin-based and SNP-based (on liability scale) heritability for different psychiatric (BIP, bipolar disorder; SCZ, schizophrenia; ADHD, attention-deficit/hyperactivity disorder; MD, major depression; ANX, generalized anxiety disorder), behavioral (AN, anorexia nervosa; AUD, alcohol use disorder; CUD, cannabis use disorder), or neurological (ASD, autism spectrum disorder; AD, Alzheimer's disorder; OCD, obsessive-compulsive disorder; TS, Tourette's syndrome) disorders. Error bars are shown for SNP-based estimates from LDSC.

Figure 2

Table 1. Summary of bipolar disorder GWAS

Figure 3

Fig. 3. Statistical power calculations for current and future GWAS. The variance explained by genome-wide significant variants (y-axis) is calculated for increasing GWAS sample sizes (x-axis) using the univariate causal mixture model (Holland et al., 2020). The legend describes the estimated GWAS sample sizes (SE) needed to capture 50% of the genetic variance (horizontal dashed line) associated with each trait. Stars indicate the sample sizes of currently available GWAS, and circles indicate the estimated sample sizes needed to capture 50% of the genetic variance for each trait. Traits include attention-deficit/hyperactivity disorder (ADHD) (Demontis et al., 2019b), autism spectrum disorder (ASD) (Grove et al., 2019), bipolar disorder (BD) (Mullins et al., 2020), depression (MDD) (Howard et al., 2019), and schizophrenia (SCZ) (Pardiñas et al., 2018). Height is included as a somatic control (no genetic correlation exists between height and bipolar disorder) (Yengo et al., 2018). s.e., standard error.