186
Views
28
CrossRef citations to date
0
Altmetric
Review

Evidence for single nucleotide polymorphisms and their association with bipolar disorder

Pages 1573-1582 | Published online: 11 Oct 2013

Abstract

Bipolar disorder (BD) is a complex disorder with a number of susceptibility genes and environmental risk factors involved in its pathogenesis. In recent years, huge progress has been made in molecular techniques for genetic studies, which have enabled identification of numerous genomic regions and genetic variants implicated in BD across populations. Despite the abundance of genetic findings, the results have often been inconsistent and not replicated for many candidate genes/single nucleotide polymorphisms (SNPs). Therefore, the aim of the review presented here is to summarize the most important data reported so far in candidate gene and genome-wide association studies. Taking into account the abundance of association data, this review focuses on the most extensively studied genes and polymorphisms reported so far for BD to present the most promising genomic regions/SNPs involved in BD. The review of association data reveals evidence for several genes (SLC6A4/5-HTT [serotonin transporter gene], BDNF [brain-derived neurotrophic factor], DAOA [D-amino acid oxidase activator], DTNBP1 [dysbindin], NRG1 [neuregulin 1], DISC1 [disrupted in schizophrenia 1]) to be crucial candidates in BD, whereas numerous genome-wide association studies conducted in BD indicate polymorphisms in two genes (CACNA1C [calcium channel, voltage-dependent, L type, alpha 1C subunit], ANK3 [ankyrin 3]) replicated for association with BD in most of these studies. Nevertheless, further studies focusing on interactions between multiple candidate genes/SNPs, as well as systems biology and pathway analyses are necessary to integrate and improve the way we analyze the currently available association data.

Introduction

Bipolar disorder (BD) is a common psychiatric illness characterized by recurrent episodes of mania and depression. Heritability, as calculated in recent twin studies, is estimated at about 85%,Citation1 which makes BD one of the most heritable multifactorial medical conditions. It is complex in nature, with underlying numerous susceptibility genes as well as environmental risk factors contributing to heterogeneity in observed clinical phenotypes. Over 20 family studies have demonstrated that the relative risk in the first-degree relatives is seven-fold greater than the risk in general population, whereas twin studies have shown a 70% concordance rate in monozygotic twins, indicating that the genetic component is very important in the development of BD. Moreover, family studies have indicated common genetic determinants with other psychiatric disorders such as schizophrenia and autism.Citation2,Citation3

In the last decades, genetic studies in BD focused on linkage analyses and association studies of candidate genes, resulting in numerous chromosomal regions and gene polymorphisms linked to the disease. However, the results so far have been inconsistent across the studies, possibly due to differences between the populations analyzed, limited sample sizes with insufficient power to detect an association, as well as different definitions and descriptions used for the diagnosis of clinical phenotype. Recently, genome-wide association studies (GWASs) have also been applied to discover novel candidates and replicate the findings from previous association analyses of single susceptibility genes.

The aim of this review is to summarize the findings from numerous association studies, focusing on variants within genes that have been widely analyzed and replicated or functionally tested for relevance to the BD phenotype.

Candidate gene studies

In this approach, a specific gene based on its function is tested for possible involvement in the pathogenesis of disease. Genetic markers of known function or located in potentially important regulatory gene regions are analyzed in case-control studies to determine if the variant is involved in disease. This method enables the detection of variants of physiological relevance using a limited number of statistical tests; however, it is confined to suspected genes with no possibility of identifying novel candidates. A large number of association studies for candidate genes in BD have been performed so far. Some of the candidate genes associated with BD are presented in .

Table 1 Candidate genes most studied in association with bipolar disorder

However, most of these studies present discrepant findings not replicated by others, which makes the interpretation of data difficult. The most consistent associations were observed for a few candidates: SLC6A4/5-HTT (serotonin transporter), BDNF (brain-derived neurotrophic factor), COMT (catechol-O-methyltransferase), DISC1 (disrupted in schizophrenia 1, coding for a neuronal growth-related protein), DTNBP1 (dysbindin), DAOA (D-amino acid oxidase activator), and NRG1 (neuregulin 1).

SLC6A4/5-HTT has been one of the most extensively studied in psychiatric diseases.Citation4 The transporter plays a crucial role in the active reuptake of serotonin at the synapse that is a known target of selective serotonin reuptake inhibitor antidepressants, which block transporter action. Although numerous studies analyzing its possible association with BD have been performed to date, most of them have focused on the variable number of tandem repeats polymorphism, which has turned out to have functional significance on the transcription initiation of the transporter. Only several association studies including single nucleotide polymorphisms (SNPs) within 5-HTT have been reported so far, but none of these has shown association with BD.Citation5Citation8

Another candidate gene extensively studied in BD is BDNF, a factor involved in brain development – in particular, in the catecholamine system implicated in BD. Its expression is increased on lithium and antidepressant administration. The first described variant includes a functional SNP, Val/Met substitution (rs6265), that affects peptide trafficking and release. Association studies of this functional polymorphism with BD produced inconsistent findings across the populations, with either positiveCitation9Citation17 or negative results.Citation18Citation23 The meta-analysis of this substitution has not confirmed the association with BD.Citation24 However, another meta-analysis by Fan and Sklar,Citation25 based on original published association studies between the Val66Met polymorphism and BD, including 14 studies (4,248 cases and 7,080 control subjects and 858 nuclear families), showed modest but significant evidence for the association of this locus with BD. Some association studies of other SNPs within BDNF (rs6265, rs1519480, rs12273363, rs11030107, rs11030104, rs11030119; haplotype consisting of rs6265 and rs988748; rs1519480, rs7127507) support their role in the pathogenesis of BD,Citation17,Citation26,Citation27 but others could not replicate the association of rs6265 and rs11030101 with BD.Citation23

COMT is an enzyme involved in monoamine degradation and its gene has been suggested as a candidate for BD.Citation28 The most studied polymorphism, Val/Met substitution, which has been shown to influence enzyme activity, has not been confirmed to be associated with BDCitation29Citation35 except in some studies.Citation36,Citation37 However, other polymorphisms (rs165599, rs2097603, Val/Met variant, rs737865) were analyzed with positive results for association with BD.Citation35,Citation38,Citation39

Some of the important candidate genes (DISC1, DTNBP1, DAOA, NRG1) with evidence for association with BD were initially identified by mapping studies in schizophrenia and later also shown to be involved in BD.

DISC1 was identified by mapping a balanced translocation between chromosome 1 and 11 that was found to segregate with both schizophrenia and BD in a large Scottish pedigree and was the causative mutation in the family.Citation40 Further studies supported the association of this gene with BD, both single markers (rs6675281; rs1538979, rs821577; 1030711; rs2492367, rs7546310) and haplotypes (rs7546310A-rs82159T; rs766288A-rs2492367C; rs1000731A-rs7546310C, rs2738864C-rs16841582C; rs1030711A-rs751229C-rs1285730A-rs3738401G),Citation41Citation46 except the study by Devon et al,Citation47 in which no association of DISC1 polymorphisms (A844G, C1460T, T2163A, and T11870C) with BD was reported.

DTNBP1 is another candidate gene that was reported initially for schizophrenia but has also shown association with BD. The protein is expressed in all neuronal populations of the hippocampus and plays a role in glutamatergic pathway signaling. The association of single polymorphisms (rs2005976, rs760761; rs2619522) and haplotypes (rs3213207A-rs1011313C-rs2005976G-rs760761T-rs2619522A; TCGG and GTAA of SNPs: rs2619522, rs760761, rs2005976, rs2619528; rs2619538-rs2619522) has been supported by several positive studies.Citation48Citation51

DAOA (G72/G30) was first identified by linkage family studies to chromosome 13q, a region also linked to BD.Citation52,Citation53 A number of studies showed positive association with BD (rs1935058; rs1341402, rs1935062, rs778294; rs947267; haplotypes: rs2111902A-rs3918346T and rs746187G-rs3916972G; haplotypes: CGCAT, TAACT, CGACT, TGCAT of five SNPs: rs1935058, rs2391191, rs1935062, rs947267, rs954581).Citation54Citation57 Further evidence was reported in a large study analyzing nine polymorphisms that tag the common genetic variations in DAOA in association with susceptibility to mood episodes in BD (rs391695, rs1341402, DAOA_3′UTR_SNP12).Citation58 However, associations of the studied DAOA polymorphisms (rs2391191G, rs778294T, rs1341402T, rs1935058C, and rs1935062C) were not confirmed in a meta-analysis by Shi et al.Citation59

NRG1 interacts with ErbB4 to regulate glutamate signaling via the N-methyl-D-aspartate receptor. The gene was first identified by linkage to chromosome 8p in Icelandic families with schizophrenia,Citation60 followed by association of this gene with BD (single markers: SNP8NRG221533; rs553950, rs327329, rs7007662; haplotypes: NP8NRG221533-rs4298458-SNP8NRG241930-SNP8NRG243177; rs2919390-rs6988339-rs3757930).Citation61,Citation62 However, this was not replicated in an Irish trios study (SNP8NRG221533, 478B14-878, 420M9-139, SNP8NRG243177).Citation63

GWASs

This method involves the simultaneous analysis of hundreds of SNPs distributed throughout the genome in large cohorts of patients and controls to identify genetic markers associated with the disease. In contrast to most candidate gene studies, GWASs are well powered and the genes found are usually replicated in other studies, demonstrating their priority over candidate gene studies. In BD, the first such study was published by the Wellcome Trust Case Control Consortium (WTCCC) in 2007. The study included about 2,000 bipolar cases and 3,000 controls and analysis of 500,000 SNPs revealed that the region with the strongest evidence for association with BD was at locus 16p12 (P=10−8), which contains three genes of potential pathological relevance to BD (PALB2 [partner and localizer of BRCA2], NDUFAB1 [NADH dehydrogenase (ubiquinone) 1], DCTN5 [dynactin 5]).Citation41 Another GWAS was performed by Sklar et alCitation64 on the sample for the Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD), a large treatment study in BD. This analysis identified two genes, MYO5B (myosin 5B) and TSPAN8 (tetraspanin-8), associated with BD, although this finding was not replicated in an independent sample.

The gene that showed evidence for association in both those studies was CACNA1C, a member of a family of L-type calcium channels. This candidate gene was also identified in a combined analysis study, by Ferreira et al,Citation65 of the two aforementioned datasets and additional samples for a total of 4,387 cases and 6,209 controls. This study also identified another candidate with the strongest evidence for association with BD: the ANK3 (ankyrin 3) gene encoding ankyrin G, a cytoskeletal protein expressed in the brain and involved in attaching sodium channels to the cytoskeleton.

A later GWAS performed by Smith et alCitation66 of a National Institute of Mental Health (NIMH) Genetics Initiative for Bipolar Disorder sample, a part of the Bipolar Genome Study (BiGS), failed to identify any SNP with a genome-wide significance; however, they showed suggestive evidence for association of the following candidate genes: NAP5 (NCK-associated protein 5), DPY19L3 (dpy-19-like 3 [Caenorhabditis elegans]), and NTRK2 (neurotrophic tyrosine kinase receptor type 2). In particular, the latter gene is a good candidate, encoding the Trkb tyrosine kinase receptor for BDNF that has been extensively studied in psychiatric disorders. The study by Smith et al is also one of the few that has examined a non-Caucasian population. Combined analysis of European-American and African-American samples enabled identification of several genes showing association in both populations, but with different alleles (ROR1 [receptor tyrosine kinase-like orphan receptor 1], RGS5 [regulator of G protein signaling 5], BTBD16 [BTB (POZ) domain containing 16]), suggesting that alternative variants in the same genes predispose to BD depending on the population.

Similar to this study, no significant evidence for association was observed in a GWAS by Scott et alCitation67 in three populations of 3,683 Caucasian cases and 14,507 controls. The SNPs with suggestive evidence for association included the following genes: ITIH1 (inter-alpha-trypsin inhibitor heavy chain 1), GNL3 (guanine nucleotide binding protein-like 3), NEK4 (NIMA-related kinase 4), and ITIH3 (inter-alpha-trypsin inhibitor heavy chain 3), as well as replication for ANK3 gene.

A later study undertaken by the Psychiatric GWAS Consortium Bipolar Disorder Working GroupCitation68 published a combined GWAS of 11,974 bipolar patients and 51,792 controls as part of the Psychiatric GWAS Consortium. In that study, genome-wide significant evidence of association for CACNA1C was confirmed and for the identification of a new intronic variant in ODZ4 gene presented.

A novel genetic variation (rs1064395) within the NCAN (neurocan) gene that showed genome-wide significant association (P=3.02×10−8) was identified in a combined GWAS and first follow-up step study performed in 2,411 BD patients and 3,613 controls.Citation69 This finding was replicated in a second follow-up step study (6,030 patients and 31,749 controls; P=2.74×10−4). The combined analysis of all study samples yielded a P-value of 2.14×10−9, providing evidence that rs1064395 is a common risk factor for BD. NCAN encodes neurocan, an extracellular matrix glycoprotein involved in cell adhesion and migration, with expression localized within cortical and hippocampal areas involved in cognition and emotion regulation implicated in BD

The most recent GWAS by Green et alCitation70 of an independent UK sample of 1,218 BD cases and 2,913 controls replicated previous findings by Ferreira et alCitation65 for two of the three most strongly associated chromosomal regions in the study, CACNA1C (rs1006737, P=4.09×10−4) and 15q14 (rs2172835, P=0.043), but not ANK3 (rs10994336, P=0.912). Moreover, the authors performed combined analysis of two populations, ImmunoChip data (569 quasi-independent SNPs from the 3,016 SNPs genotyped) with the recently published Psychiatric Genome-Wide Association Study Consortium Bipolar Disorder Working Group (PGC-BD) meta-analysis data and found two novel variants associated with BD: rs7296288 (P=8.97×10−9), an intergenic polymorphism on chromosome 12 located between RHEBL1 (Ras homolog enriched in brain like 1) and DHH (desert hedgehog), and rs3818253 (P=3.88×10−8), an intronic SNP on chromosome 20q11.2 in the TRPC4AP (transient receptor potential cation channel, subfamily C, member 4 associated protein) gene, which lies in a high linkage disequilibrium region along with the genes GSS (glutathione synthetase) and MYH7B (myosin heavy chain 7B).

A few national GWASs have also been performed. Among others, one in a Bulgarian populationCitation71 and two in Scandinavian populations – one SwedishCitation72 and the other Norwegian – followed by replication in an Icelandic population;Citation73 all of these failed to identify any locus of genome-wide significance for association with BD. However, in the Bulgarian study, the authors mentioned three variants with possible involvement in BD: rs8099939 (P=2.12×10−6) in the GRIK5 (glutamate receptor, ionotropic, kainate 5) gene, rs6122972 (P=3.11×10−6) in the PARD6B (par-6 family cell polarity regulator beta) gene, and rs2289700 (P=9.14×10−6) in the CTSH (cathepsin H) gene.Citation71

Two GWASs in Asian populations have been performed: one by Hattori et alCitation74 in a Japanese population of 107 cases and 107 controls, and the other by Lee et alCitation75 in a Han Chinese population of 1,000 bipolar I patients and 1,000 controls. In both studies, none of the SNPs was associated with BD at genome-wide significance. A summary of GWASs is presented in .

Table 2 Genome-wide association studies (GWASs) in bipolar disorder

Recently, the Cross-Disorder Group of the Psychiatric Genomics Consortium published GWAS results for five major psychiatric disorders – autism spectrum disorder, attention deficit-hyperactivity disorder, BD, major depressive disorder, and schizophrenia – to identify loci shared between those disorders.Citation76 The analysis of 33,332 cases and 27,888 controls revealed that genome-wide significance (P<5×10−8) was reached for regions on chromosomes 3p21 and 10q24 and SNPs within two L-type voltage-gated calcium-channel subunits, CACNA1C and CACNB2 (calcium channel, voltage-dependent, beta 2 subunit), indicating that specific SNPs are associated with a range of psychiatric disorders and that variation in calcium-channel activity genes may exert pleiotropic effects on the psychopathology of psychiatric genetics.

The results from GWASs provide numerous SNPs evidenced for association with BD, with the strongest confirmation of two loci, CACNA1C and ANK3. However, lack of consistent findings throughout the studies, possibly due to insufficient power to detect associations with genes of small effects, impede drawing conclusions. Recently, a meta-analysis of GWASs and all published candidate gene-association studies of BD (a total of 487 articles) was published to evaluate the cumulative evidence of associations observed so far.Citation77 Polymorphisms in BDNF, DRD4, DAOA, and TPH1 (tryptophan hydroxylase 1) were found to be nominally significant; however, none of these findings was significant after correction for multiple testing. Moreover, none of these polymorphisms was significant in the Psychiatric GWAS Consortium Bipolar Disorder studyCitation76

Phenotypes related to BD

Taking into account the genetic heterogeneity of BD, it has been suggested that clinical phenotypes may be more useful in delineating the genetic variants contributing to BD than standard diagnostic models.Citation78 Clinical sub-phenotypes of BD may identify more homogeneous subsets of patients who can be studied with increased power to detect genetic variation. To date, several studies have applied this concept to data from GWASs.

In a GWAS of BD patients with seasonal patterned mania (seasonal or non-seasonal patterned manic episodes), the most significant association was observed for rs41350144, which lies within an intron of the NF1A (nuclear factor I/A) gene on 1p31 (P=3.08×10−7), suggesting it may predispose to this subtype of BD.Citation79

A GWAS of mood-incongruent psychotic features in bipolar patients (2,196 cases with mood-incongruent psychotic features and 8,148 controls) revealed no association of genome-wide significance; however, several regions with suggestive evidence of association (P<10−6) were found: 6q14.2 within the PRSS35 (protease serine 35)/SNAP91 (synaptosomal-associated protein) gene complex (rs1171113, P=9.67×10−8), 3p22.2 downstream of TRANK/LBA1 (tetratricopeptide repeat and ankyrin repeat containing 1) (rs9834970, P=9.71×10−8), and 14q24.2 in an intron of numb homolog (Drosophila) (rs2333194, P=7.03×10−7).Citation80

In a GWAS of factor dimensions in 927 clinically well-characterized BD patients of German ancestry, the authors found that the factor dimension “negative mood delusions” was significantly associated with one variant (rs9875793; n=927; P=4.65×10−8).Citation81 This SNP is located in an intergenic region of 3q26.1 in the proximity of the SLC2A2 (solute carrier family 2 [facilitated glucose transporter], member 2) gene. In case-control analyses, significant association with the G allele of rs9875793 was only observed in the subgroup of 89 BD patients who displayed symptoms of negative mood delusions. Further support for the association of rs9875793 with BD in patients displaying the negative mood delusions symptom was obtained from an European-American sample of 1,247 BD patients and 1,434 controls.

The study that first tested bipolar patients according to age-at-onset (AAO) subgroups was performed by Dizier et alCitation82 in a sample of 443 unrelated bipolar patients and 1,731 controls. The study provided evidence for genetic variation within the ADRB2 (β2-adrenoreceptor) gene region that is specifically associated with the early onset form of BD. However, this finding could not be replicated in the WTCCC sample due to poor genotyping coverage of the ADRB2 gene (the SNPs that showed the strongest signal for association in the study were not available in the WTCCC sample and were not in LD (linkage disequilibrium) with the SNPs available in the replication sample). Therefore, the finding requires further investigation.

A meta-analysis of two widely studied sub-phenotypes of BD, AAO and psychotic symptoms, which are familial and clinically significant, was conducted by Belmonte Mahon et alCitation83 on combined data from three GWASs: the NIMH Bipolar Disorder Genetic Association Information Network (GAIN-BP), NIMH BiGS, and a German sample, with a total of 2,836 BD cases with information on sub-phenotypes and 2,744 controls. No SNP reached genome-wide significance for either sub-phenotype. The same results were observed in a meta-analysis with an independent replication sample. This indicates that AAO and psychotic symptoms in BD may be influenced by other variants not measured well by SNP arrays, such as rare alleles.

Family and twin studies suggest that susceptibility for suicide attempts is heritable and distinct from mood disorder susceptibility, and the high resolution of the GWAS approach facilitates the detection of risk loci. The first GWAS performed for lifetime suicide attempts sub-phenotype was conducted by Perlis et al.Citation84 BD subjects were drawn from the Systematic Treatment Enhancement Program for Bipolar Disorder cohort, the WTCCC bipolar cohort, and the University College London cohort. Replication was pursued in the NIMH Genetic Association Information Network BD project and a German clinical cohort. The strongest evidence of association for suicide attempts in BD was observed in a region without identified genes (rs1466846) and five loci showed suggestive evidence of association.

Another attempted suicide GWAS that compared the SNP genotypes of 1,201 BD subjects with a history of suicide attempts to the genotypes of 1,497 BD subjects without a history of suicide attempts was performed by Willour et al.Citation85 The authors found 2,507 SNPs with evidence for association with suicide attempts at P<0.001, but these associations were not significantly associated in the replication sample after correcting for multiple testing. However, the combined analysis of the two sample sets produced an association signal on 2p25 (rs300774) at the threshold of genome-wide significance (P=5.07×10−8). This variant is located in a large linkage disequilibrium block containing the ACP1 (acid phosphatase 1) gene, the expression of which is significantly elevated in BD subjects who have completed suicide. The results of both GWASs suggest that inherited risk for suicide among bipolar patients is unlikely to be the result of individual common variants of large effects.

A GWAS testing comorbidities in BD (BD with psychosis and/or substance abuse in the absence of alcohol dependence) showed association with the rare variant (rs1039002) in the vicinity of the PDE10A (phosphodiesterase 10A) gene (P=1.7×10−8), which was implicated in the pathophysiology of psychosis. Another rare variant, rs12563333 on chromosome 1q41 close to the MARK1 (MAP/microtubule affinity-regulating kinase 1) gene, demonstrated an almost genome-wide level of significance in this subgroup (P=5.9×10−8). Homozygotes for the minor allele were present in cases and absent in controls. BD with alcohol dependence and other comorbidities was associated with SNP rs2727943 (P=3.3×10−8) on chromosome 3p26.3 located between the genes BIG-2 (contactin-4 precursor) and CNTN6 (contactin 6). BD with low probability of comorbid conditions did not show significant associations.Citation86

Neuroimaging is commonly used to characterize brain activity that is altered in psychiatric disorders including BD By use of functional magnetic resonance imaging, it was found that amygdala activation during a face-processing task differs between bipolar patients and healthy controls.Citation87 In a GWAS, a SNP (rs2023454) in DOK5 (docking protein 5) gene involved in the neurotrophin signaling pathway was found to be associated with right amygdala activation.Citation88 Another GWAS performed on neurocognition in 157 BD patients and 353 controlsCitation89 identified three intronic SNPs in the PTPRO (protein tyrosine phosphatase receptor type O) gene associated with learning and memory (rs17222089, rs11056571, and rs2300290) and rs719714 near WDR72 (WD repeat domain 72) associated with executive functioning as well as highly significant interaction between FOXQ1 (forkhead box Q1) and SUMO1P1 (SUMO1 pseudogene 1) SNPs for psychomotor speed.

Several studies analyzing the influence of variation in candidate genes for BD on neuroanatomy and neurocognition have also been performed. A SNP in the CACNA1C gene (rs1006737) previously associated with BD in several GWASs, was also found related to alterations in structural and functional magnetic resonance imaging, with A allele associated with increased gray-matter volume and reduced functional connectivity within the cortico-limbic frontotemporal neural system,Citation90 as well as worsened performance of executive function tests in BD patients.Citation91 However, it was not associated with spatial working memory in BD patients.Citation92 Evidence for the involvement of SNPs within the DISC1 gene, another strong candidate for BD, in neuroanatomy and neurocognition was provided by Carless et al.Citation93 Recently, a study by Li et alCitation94 analyzing SNPs in CREB1 (cAMP responsive element binding protein 1) gene in BD patients of European ancestry, reported a significant association of risk SNPs with a decreased hippocampal volume and diminished activation of the left hippocampus, further suggesting their involvement in BD susceptibility.

Conclusion

The results from candidate gene association studies and GWASs have provided some findings for BD and indicated a number of genes and variants involved. However, inconsistency between studies, as well as only modest replication of variants identified for BD by independent analyses – in particular, in the case of candidate gene studies – impede drawing conclusions. The discrepancies in candidate gene studies may partly result from the clinical and genetic heterogeneity of BD and its complex inheritance model but also from methodological issues such as the definition of clinical phenotype.

Further, it should also be taken into account that although many genes/risk alleles are implicated in BD, the individual effect sizes of each allele contribute to only a small fraction (∼3%) of the total population variance.Citation95 Despite the high estimated heritability of BD, the thousands of markers analyzed by GWASs explain only about 5% of phenotypic variance, despite studies of tens of thousands of people. As such, much of the heritability of BD has been missed by GWAS findings so far. The possible explanations of this missing heritability include analysis of rare variants (according to the “common disease-multiple rare variants” hypothesis); analysis of structural genomic variants, termed “copy number variants,” with higher odds ratios than common variants (SNPs); as well as the functional characterization of specific SNPs, such as their effects on gene expression or DNA methylation. Next-generation sequencing offers help in elucidating if the missing heritability is due to missed genetic associations. Moreover, taking into account the polygenic inheritance of BD, with a large number of markers with small individual effects collectively accounting for disease risk, novel approaches including systems biology and pathway analysis may be helpful in identifying functional interactions between multiple genes involved in the pathophysiology of BD. These methods are already being used in ongoing experimentsCitation96Citation98 to determine the underlying mechanisms and identify causal variants for this disease.

Thus, the genetic findings in BD, although promising, are far from being translated into clinical practice.

Disclosure

The author declares no conflicts of interest in this work.

References

  • BienvenuOJDavydowDSKendlerKSPsychiatric ‘diseases’ versus behavioral disorders and degree of genetic influencePsychol Med2011411334020459884
  • LichtensteinPYipBHBjörkCCommon genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based studyLancet2009373965923423919150704
  • SullivanPFMagnussonCReichenbergAFamily history of schizophrenia and bipolar disorder as risk factors for autismArch Gen Psychiatry201269111099110322752149
  • CaspiAHaririARHolmesAUherRMoffittTEGenetic sensitivity to the environment: the case of the serotonin transporter gene and its implications for studying complex diseases and traitsAm J Psychiatry2010167550952720231323
  • SunHSWangHCLaiTJWangTJLiCMSequence variants and haplotype analysis of serotonin transporter gene and association with bipolar affective disorder in TaiwanPharmacogenetics200414317317915167705
  • IkedaMIwataNSuzukiTNo association of serotonin transporter gene (SLC6A4) with schizophrenia and bipolar disorder in Japanese patients: association analysis based on linkage disequilibriumJ Neural Transm2006113789990516082508
  • MansourHATalkowskiMEWoodJSerotonin gene polymorphisms and bipolar I disorder: focus on the serotonin transporterAnn Med200537859060216338761
  • AlaertsMCeulemansSForeroDDetailed analysis of the serotonin transporter gene (SLC6A4) shows no association with bipolar disorder in the Northern Swedish populationAm J Med Genet B Neuropsychiatr Genet2009150B458559218792946
  • SklarPGabrielSBMcInnisMGFamily-based association study of 76 candidate genes in bipolar disorder: BDNF is a potential risk locus. Brain-derived neutrophic factorMol Psychiatry20027657959312140781
  • Neves-PereiraMMundoEMugliaPKingNMacciardiFKennedyJLThe brain-derived neurotrophic factor gene confers susceptibility to bipolar disorder: evidence from a family-based association studyAm J Hum Genet200271365165512161822
  • LohoffFWSanderTFerraroTNDahlJPGallinatJBerrettiniWHConfirmation of association between the Val66Met polymorphism in the brain-derived neurotrophic factor (BDNF) gene and bipolar I disorderAm J Med Genet B Neuropsychiatr Genet2005139B1515316152572
  • GreenEKRaybouldRMacgregorSGenetic variation of brain-derived neurotrophic factor (BDNF) in bipolar disorder: case-control study of over 3000 individuals from the UKBr J Psychiatry2006188212516388065
  • OkadaTHashimotoRNumakawaTA complex polymorphic region in the brain-derived neurotrophic factor (BDNF) gene confers susceptibility to bipolar disorder and affects transcriptional activityMol Psychiatry200611769570316568151
  • KremeyerBHerzbergIGarciaJTransmission distortion of BDNF variants to bipolar disorder type I patients from a South American population isolateAm J Med Genet B Neuropsychiatr Genet2006141B543543916741941
  • XuJLiuYWangPPositive association between the brain-derived neurotrophic factor (BDNF) gene and bipolar disorder in the Han Chinese populationAm J Med Genet B Neuropsychiatr Genet2010153B127527919330778
  • WangZLiZChenJAssociation of BDNF gene polymorphism with bipolar disorders in Han Chinese populationGenes Brain Behav201211552452822548711
  • SearsCMarkieDOldsRFitchesAEvidence of associations between bipolar disorder and the brain-derived neurotrophic factor (BDNF) geneBipolar Disord2011137–863063722085476
  • NakataKUjikeHSakaiAAssociation study of the brain-derived neurotrophic factor (BDNF) gene with bipolar disorderNeurosci Lett20033371172012524161
  • HongCJLiuHCLiuTYLinCHChengCYTsaiSJBrain-derived neurotrophic factor (BDNF) Val66Met polymorphisms in Parkinson’s disease and age of onsetNeurosci Lett20033531757714642442
  • KunugiHUekiAOtsukaMA novel polymorphism of the brain-derived neurotrophic factor (BDNF) gene associated with late-onset Alzheimer’s diseaseMol Psychiatry200161838611244490
  • SkibinskaMHauserJCzerskiPMAssociation analysis of brain-derived neurotrophic factor (BDNF) gene Val66Met polymorphism in schizophrenia and bipolar affective disorderWorld J Biol Psychiatry20045421522015543516
  • Neves-PereiraMCheungJKPasdarABDNF gene is a risk factor for schizophrenia in a Scottish populationMol Psychiatry200510220821215630410
  • YeCYXuYQHuHAn association study of brain-derived neurotrophic factor gene polymorphism in bipolar disordersZhonghua Yi Xue Za Zhi2009892718971901 Chinese19953912
  • KanazawaTGlattSJKia-KeatingBYonedaHTsuangMTMeta-analysis reveals no association of the Val66Met polymorphism of brain-derived neurotrophic factor with either schizophrenia or bipolar disorderPsychiatr Genet200717316517017417060
  • FanJSklarPGenetics of bipolar disorder: focus on BDNF Val66Met polymorphismNovartis Found Symp20082896072 discussion 72–73, 87–9318497095
  • SchumacherJJamraRABeckerTEvidence for a relationship between genetic variants at the brain-derived neurotrophic factor (BDNF) locus and major depressionBiol Psychiatry200558430731416005437
  • LiuLForoudTXueiXEvidence of association between brain-derived neurotrophic factor gene and bipolar disorderPsychiatr Genet200818626727419018231
  • CraddockNOwenMJO’DonovanMCThe catechol-O-methyl transferase (COMT) gene as a candidate for psychiatric phenotypes: evidence and lessonsMol Psychiatry200611544645816505837
  • GutiérrezBBertranpetitJGuillamatRAssociation analysis of the catechol O-methyltransferase gene and bipolar affective disorderAm J Psychiatry199715411131158988970
  • No association between bipolar disorder and alleles at a functional polymorphism in the COMT geneBiomed European Bipolar Collaborative GroupBr J Psychiatry19971705265289330018
  • LachmanHMKelsoeJMorenoLKatzSPapolosDFLack of association of catechol-O-methyltransferase (COMT) functional polymorphism in bipolar affective disorderPsychiatr Genet19977113179264133
  • KirovGJonesIMcCandlessFCraddockNOwenMJFamily-based association studies of bipolar disorder with candidate genes involved in dopamine neurotransmission: DBH, DAT1, COMT, DRD2, DRD3 and DRD5Mol Psychiatry19994655856510578238
  • KunugiHValladaHPHodaFNo evidence for an association of affective disorders with high- or low-activity allele of catechol-o-methyltransferase geneBiol Psychiatry19974242822859270905
  • SerrettiARotondoALorenziCSmeraldiECassanoGBCatechol-O-methyltransferase gene variants in mood disorders in the Italian populationPsychiatr Genet200616518118216969269
  • BurdickKEFunkeBGoldbergJFCOMT genotype increases risk for bipolar I disorder and influences neurocognitive performanceBipolar Disord20079437037617547583
  • LiTValladaHCurtisDCatechol-O-methyltransferase Val158Met polymorphism: frequency analysis in Han Chinese subjects and allelic association of the low activity allele with bipolar affective disorderPharmacogenetics1997753493539352569
  • Mynett-JohnsonLAMurphyVEClaffeyEShieldsDCMcKeonPPreliminary evidence of an association between bipolar disorder in females and the catechol-O-methyltransferase genePsychiatr Genet1998842212259861640
  • ShifmanSBronsteinMSternfeldMCOMT: a common susceptibility gene in bipolar disorder and schizophreniaAm J Med Genet B Neuropsychiatr Genet2004128B1616415211633
  • FunkeBMalhotraAKFinnCTCOMT genetic variation confers risk for psychotic and affective disorders: a case control studyBehav Brain Funct200511916232322
  • BlackwoodDHFordyceAWalkerMTSt ClairDMPorteousDJMuirWJSchizophrenia and affective disorders – cosegregation with a translocation at chromosome 1q42 that directly disrupts brain-expressed genes: clinical and P300 findings in a familyAm J Hum Genet200169242843311443544
  • Wellcome Trust Case Control ConsortiumGenome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controlsNature2007447714566167817554300
  • HodgkinsonCAGoldmanDJaegerJDisrupted in schizophrenia 1 (DISC1): association with schizophrenia, schizoaffective disorder, and bipolar disorderAm J Hum Genet200475586287215386212
  • ThomsonPAWrayNRMillarJKAssociation between the TRAX/DISC locus and both bipolar disorder and schizophrenia in the Scottish populationMol Psychiatry2005107657668 61615838535
  • HennahWThomsonPMcQuillinADISC1 association, heterogeneity and interplay in schizophrenia and bipolar disorderMol Psychiatry200914986587318317464
  • SchosserAGaysinaDCohen-WoodsSAssociation of DISC1 and TSNAX genes and affective disorders in the depression case-control (DeCC) and bipolar affective case-control (BACCS) studiesMol Psychiatry201015884484919255581
  • XiaoYZhangJWangYLimited association between Disrupted in Schizophrenia 1 (DISC1) gene and bipolar disorder in the Chinese populationPsychiatr Genet2011211424621222298
  • DevonRSAndersonSTeaguePWIdentification of polymorphisms within Disrupted in Schizophrenia 1 and Disrupted in Schizophrenia 2, and an investigation of their association with schizophrenia and bipolar affective disorderPsychiatr Genet2001112717811525420
  • RaybouldRGreenEKMacGregorSBipolar disorder and polymorphisms in the dysbindin gene (DTNBP1)Biol Psychiatry200557769670115820225
  • BreenGPrataDOsborneSAssociation of the dysbindin gene with bipolar affective disorderAm J Psychiatry200616391636163816946192
  • PaeCUSerrettiAMandelliLEffect of 5-haplotype of dysbindin gene (DTNBP1) polymorphisms for the susceptibility to bipolar I disorderAm J Med Genet B Neuropsychiatr Genet2007144B570170317192893
  • JooEJLeeKYJeongSHDysbindin gene variants are associated with bipolar I disorder in a Korean populationNeurosci Lett2007418327227517433541
  • Detera-WadleighSDBadnerJABerrettiniWHA high-density genome scan detects evidence for a bipolar-disorder susceptibility locus on 13q32 and other potential loci on 1q32 and 18p11.2Proc Natl Acad Sci U S A199996105604560910318931
  • ShawSHMroczkowski-ParkerZShekhtmanTLinkage of a bipolar disorder susceptibility locus to human chromosome 13q32 in a new pedigree seriesMol Psychiatry20038555856412808435
  • HattoriELiuCBadnerJAPolymorphisms at the G72/G30 gene locus, on 13q33, are associated with bipolar disorder in two independent pedigree seriesAm J Hum Genet20037251131114012647258
  • SchumacherJJamraRAFreudenbergJExamination of G72 and D-amino-acid oxidase as genetic risk factors for schizophrenia and bipolar affective disorderMol Psychiatry20049220320714966479
  • ChenYSAkulaNDetera-WadleighSDFindings in an independent sample support an association between bipolar affective disorder and the G72/G30 locus on chromosome 13q33Mol Psychiatry2004918792 image 514699445
  • PrataDBreenGOsborneSMunroJSt ClairDCollierDAssociation of DAO and G72(DAOA)/G30 genes with bipolar affective disorderAm J Med Genet B Neuropsychiatr Genet2008147B691491718165970
  • WilliamsNMGreenEKMacgregorSVariation at the DAOA/G30 locus influences susceptibility to major mood episodes but not psychosis in schizophrenia and bipolar disorderArch Gen Psychiatry200663436637316585465
  • ShiJBadnerJAGershonESLiuCAllelic association of G72/G30 with schizophrenia and bipolar disorder: a comprehensive meta-analysisSchizophr Res2008981–3899718023149
  • StefanssonHSarginsonJKongAAssociation of neuregulin 1 with schizophrenia confirmed in a Scottish populationAm J Hum Genet2003721838712478479
  • GreenEKRaybouldRMacgregorSOperation of the schizophrenia susceptibility gene, neuregulin 1, across traditional diagnostic boundaries to increase risk for bipolar disorderArch Gen Psychiatry200562664264815939841
  • ThomsonPAChristoforouAMorrisSWAssociation of Neuregulin 1 with schizophrenia and bipolar disorder in a second cohort from the Scottish populationMol Psychiatry20071219410416940976
  • CassidyFRocheSClaffeyEMcKeonPFirst family-based test for association of neuregulin with bipolar affective disorderMol Psychiatry200611870670716868568
  • SklarPSmollerJWFanJWhole-genome association study of bipolar disorderMol Psychiatry200813655856918317468
  • FerreiraMAO’DonovanMCMengYAWellcome Trust Case Control ConsortiumCollaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorderNat Genet20084091056105818711365
  • SmithENBlossCSBadnerJAGenome-wide association study of bipolar disorder in European American and African American individualsMol Psychiatry200914875576319488044
  • ScottLJMugliaPKongXQGenome-wide association and meta-analysis of bipolar disorder in individuals of European ancestryProc Natl Acad Sci U S A2009106187501750619416921
  • Psychiatric GWAS Consortium Bipolar Disorder Working GroupLarge-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4Nat Genet2011431097798321926972
  • CichonSMühleisenTWDegenhardtFABipolar Disorder Genome Study (BiGS) ConsortiumHamshereMO’DonovanMCGenome-wide association study identifies genetic variation in neurocan as a susceptibility factor for bipolar disorderAm J Hum Genet201188337238121353194
  • GreenEKHamshereMFortyLWTCCCJonesIRCraddockNReplication of bipolar disorder susceptibility alleles and identification of two novel genome-wide significant associations in a new bipolar disorder case-control sampleMol Psychiatry Epub10162012
  • YosifovaAMushirodaTKuboMGenome-wide association study on bipolar disorder in the Bulgarian populationGenes Brain Behav201110778979721771265
  • BergenSEO’DushlaineCTRipkeSGenome-wide association study in a Swedish population yields support for greater CNV and MHC involvement in schizophrenia compared with bipolar disorderMol Psychiatry201217988088622688191
  • DjurovicSGustafssonOMattingsdalMA genome-wide association study of bipolar disorder in Norwegian individuals, followed by replication in Icelandic sampleJ Affect Disord20101261–231231620451256
  • HattoriEToyotaTIshitsukaYPreliminary genome-wide association study of bipolar disorder in the Japanese populationAm J Med Genet B Neuropsychiatr Genet2009150B81110111719259986
  • LeeMTChenCHLeeCSGenome-wide association study of bipolar I disorder in the Han Chinese populationMol Psychiatry201116554855620386566
  • Cross-Disorder Group of the Psychiatric GenomicsCSmollerJWCraddockNIdentification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysisLancet201338198751371137923453885
  • SeifuddinFMahonPBJudyJMeta-analysis of genetic association studies on bipolar disorderAm J Med Genet B Neuropsychiatr Genet2012159B550851822573399
  • SaundersEHScottLJMcInnisMGBurmeisterMFamiliality and diagnostic patterns of subphenotypes in the National Institutes of Mental Health bipolar sampleAm J Med Genet B Neuropsychiatr Genet2008147B182617525972
  • LeeHJWooHGGreenwoodTAKripkeDFKelsoeJRA genome-wide association study of seasonal pattern mania identifies NF1A as a possible susceptibility gene for bipolar disorderJ Affect Disord2013145220020722925353
  • GoesFSHamshereMLSeifuddinFGenome-wide association of mood-incongruent psychotic bipolar disorderTransl Psychiatry20122e18023092984
  • MeierSMattheisenMVassosEBipolar Disorder Genome Study (BiGS) ConsortiumNöthenMMCichonSGenome-wide significant association between a ‘negative mood delusions’ dimension in bipolar disorder and genetic variation on chromosome 3q26.1Transl Psychiatry20122e16523010768
  • DizierMHEtainBLajnefMGenetic heterogeneity according to age at onset in bipolar disorder: a combined positional cloning and candidate gene approachAm J Med Genet B Neuropsychiatr Genet2012159B665365922628130
  • Belmonte MahonPPiroozniaMGoesFSBipolar Genome Study (BiGS) Consortium, The Wellcome Trust Case Control Consortium Bipolar Disorder GroupDepauloJRJrGenome-wide association analysis of age at onset and psychotic symptoms in bipolar disorderAm J Med Genet B Neuropsychiatr Genet2011156B337037821305692
  • PerlisRHHuangJPurcellSWellcome Trust Case Control Consortium Bipolar Disorder GroupCraddockNSklarPSmollerJWGenome-wide association study of suicide attempts in mood disorder patientsAm J Psychiatry2010167121499150721041247
  • WillourVLSeifuddinFMahonPBA genome-wide association study of attempted suicideMol Psychiatry201217443344421423239
  • KernerBLambertCGMuthénBOGenome-wide association study in bipolar patients stratified by co-morbidityPLoS One2011612e2847722205951
  • PhillipsMLLadouceurCDDrevetsWCA neural model of voluntary and automatic emotion regulation: implications for understanding the pathophysiology and neurodevelopment of bipolar disorderMol Psychiatry200813982983385718574483
  • LiuXAkulaNSkupMBrotmanMALeibenluftEMcMahonFJA genome-wide association study of amygdala activation in youths with and without bipolar disorderJ Am Acad Child Adolesc Psychiatry2010491334120215924
  • LeBlancMKulleBSundetKGenome-wide study identifies PTPRO and WDR72 and FOXQ1-SUMO1P1 interaction associated with neurocognitive functionJ Psychiatr Res201246227127822126837
  • WangFMcIntoshAMHeYGelernterJBlumbergHPThe association of genetic variation in CACNA1C with structure and function of a frontotemporal systemBipolar Disord2011137–869670022085483
  • Soeiro-de-SouzaMGBioDSDiasVVVietaEMachado-VieiraRMorenoRAThe CACNA1C risk allele selectively impacts on executive function in bipolar type I disorderActa Psychiatr Scand Epub2142013
  • ZhangQShenQXuZThe effects of CACNA1C gene polymorphism on spatial working memory in both healthy controls and patients with schizophrenia or bipolar disorderNeuropsychopharmacology201237367768422012475
  • CarlessMAGlahnDCJohnsonMPImpact of DISC1 variation on neuroanatomical and neurocognitive phenotypesMol Psychiatry2011161110961104 106321483430
  • LiMLuoXJRietschelMMooDS Bipolar ConsortiumStroh-maierJBreuerRThe Swedish Bipolar Study GroupBacklundLFrisénLThe Alzheimer’s Disease Neuroimaging InitiativeENIGMA Consortium; CHARGE ConsortiumSuBAllelic differences between Europeans and Chinese for CREB1 SNPs and their implications in gene expression regulation, hippocampal structure and function, and bipolar disorder susceptibilityMol Psychiatry2013 Epub
  • International SchizophreniaCPurcellSMWrayNRCommon polygenic variation contributes to risk of schizophrenia and bipolar disorderNature2009460725674875219571811
  • SchulzeTGAkulaNBreuerRThe Bipolar Genome StudyMcMahonFJMolecular genetic overlap in bipolar disorder, schizophrenia, and major depressive disorderWorld J Biol Psychiatry Epub392012
  • WhalleyHCPapmeyerMSprootenEThe influence of polygenic risk for bipolar disorder on neural activation assessed using fMRITransl Psychiatry20122e13022760554
  • WhalleyHCSprootenEHackettSPolygenic risk and white matter integrity in individuals at high risk of mood disorderBiol Psychiatry201374428028623453289