Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Transl Psychiatry ; 13(1): 366, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38030607

ABSTRACT

A growing body of literature recognizes associations between eating disorders (EDs) and schizophrenia and suggests that familial liability to schizophrenia in individuals with anorexia nervosa (AN) reveals distinct patterns of clinical outcomes. To further investigate the influence of schizophrenia genetic liability among individuals with EDs, we evaluated the associations between schizophrenia polygenic risk scores (PRS) and clinical presentations of individuals with EDs including their overall health condition and ED-related symptoms. Using data from two previous studies of the genetics of EDs comprising 3,573 Anorexia Nervosa Genetics Initiative (ANGI) cases and 696 Binge Eating Genetics Initiative (BEGIN) cases born after 1973 and linked to the Swedish National Patient Register, we examined the association of schizophrenia PRS on ED clinical features, psychiatric comorbidities, and somatic and mental health burden. Among ANGI cases, higher schizophrenia PRS was statistically significantly associated with higher risk of major depressive disorder (MDD) measured by hazard ratio (HR) with 95% confidence interval (CI) (HR [95% CI]: 1.07 [1.02, 1.13]) and substance abuse disorder (SUD) (HR [95% CI]: 1.14 [1.03, 1.25]) after applying multiple testing correction. Additionally, higher schizophrenia PRS was associated with decreased clinical impairment assessment scores (-0.56, 95% CI: [-1.04, -0.08]) at the conventional significance level (p < 0.05). Further, in BEGIN cases, higher schizophrenia PRS was statistically significantly associated with earlier age at first ED symptom (-0.35 year, 95% CI: [-0.64, -0.06]), higher ED symptom scores (0.16, 95% CI: [0.04, 0.29]), higher risk of MDD (HR [95% CI]: 1.18 [1.04, 1.34]) and SUD (HR [95% CI]: 1.36 [1.07, 1.73]). Similar, but attenuated, patterns held in the subgroup of exclusively AN vs other eating disorder (OED) cases. These results suggest a similar pattern of influence of schizophrenia PRS for AN and OED cases in terms of psychiatric comorbidities, but a different pattern in terms of ED-related clinical features. The disparity of the effect of schizophrenia PRS on AN vs OED merits further investigation.


Subject(s)
Anorexia Nervosa , Depressive Disorder, Major , Feeding and Eating Disorders , Schizophrenia , Substance-Related Disorders , Humans , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/epidemiology , Depressive Disorder, Major/genetics , Schizophrenia/diagnosis , Schizophrenia/epidemiology , Schizophrenia/genetics , Feeding and Eating Disorders/epidemiology , Feeding and Eating Disorders/genetics , Risk Factors , Anorexia Nervosa/epidemiology , Anorexia Nervosa/genetics , Multifactorial Inheritance
2.
Addict Biol ; 26(1): e12880, 2021 01.
Article in English | MEDLINE | ID: mdl-32064741

ABSTRACT

Eating disorders and substance use disorders frequently co-occur. Twin studies reveal shared genetic variance between liabilities to eating disorders and substance use, with the strongest associations between symptoms of bulimia nervosa and problem alcohol use (genetic correlation [rg ], twin-based = 0.23-0.53). We estimated the genetic correlation between eating disorder and substance use and disorder phenotypes using data from genome-wide association studies (GWAS). Four eating disorder phenotypes (anorexia nervosa [AN], AN with binge eating, AN without binge eating, and a bulimia nervosa factor score), and eight substance-use-related phenotypes (drinks per week, alcohol use disorder [AUD], smoking initiation, current smoking, cigarettes per day, nicotine dependence, cannabis initiation, and cannabis use disorder) from eight studies were included. Significant genetic correlations were adjusted for variants associated with major depressive disorder and schizophrenia. Total study sample sizes per phenotype ranged from ~2400 to ~537 000 individuals. We used linkage disequilibrium score regression to calculate single nucleotide polymorphism-based genetic correlations between eating disorder- and substance-use-related phenotypes. Significant positive genetic associations emerged between AUD and AN (rg = 0.18; false discovery rate q = 0.0006), cannabis initiation and AN (rg = 0.23; q < 0.0001), and cannabis initiation and AN with binge eating (rg = 0.27; q = 0.0016). Conversely, significant negative genetic correlations were observed between three nondiagnostic smoking phenotypes (smoking initiation, current smoking, and cigarettes per day) and AN without binge eating (rgs = -0.19 to -0.23; qs < 0.04). The genetic correlation between AUD and AN was no longer significant after co-varying for major depressive disorder loci. The patterns of association between eating disorder- and substance-use-related phenotypes highlights the potentially complex and substance-specific relationships among these behaviors.


Subject(s)
Feeding and Eating Disorders/genetics , Substance-Related Disorders/genetics , Alcoholism/genetics , Depressive Disorder, Major/genetics , Genome-Wide Association Study , Humans , Linkage Disequilibrium , Phenotype , Polymorphism, Single Nucleotide , Risk Factors , Schizophrenia/genetics , Tobacco Use Disorder/genetics
3.
BMC Psychiatry ; 20(1): 507, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33054774

ABSTRACT

BACKGROUND: Anorexia nervosa (AN) is a severe disorder, for which genetic evidence suggests psychiatric as well as metabolic origins. AN has high somatic and psychiatric comorbidities, broad impact on quality of life, and elevated mortality. Risk factor studies of AN have focused on differences between acutely ill and recovered individuals. Such comparisons often yield ambiguous conclusions, as alterations could reflect different effects depending on the comparison. Whereas differences found in acutely ill patients could reflect state effects that are due to acute starvation or acute disease-specific factors, they could also reflect underlying traits. Observations in recovered individuals could reflect either an underlying trait or a "scar" due to lasting effects of sustained undernutrition and illness. The co-twin control design (i.e., monozygotic [MZ] twins who are discordant for AN and MZ concordant control twin pairs) affords at least partial disambiguation of these effects. METHODS: Comprehensive Risk Evaluation for Anorexia nervosa in Twins (CREAT) will be the largest and most comprehensive investigation of twins who are discordant for AN to date. CREAT utilizes a co-twin control design that includes endocrinological, neurocognitive, neuroimaging, genomic, and multi-omic approaches coupled with an experimental component that explores the impact of an overnight fast on most measured parameters. DISCUSSION: The multimodal longitudinal twin assessment of the CREAT study will help to disambiguate state, trait, and "scar" effects, and thereby enable a deeper understanding of the contribution of genetics, epigenetics, cognitive functions, brain structure and function, metabolism, endocrinology, microbiology, and immunology to the etiology and maintenance of AN.


Subject(s)
Anorexia Nervosa , Twins, Monozygotic , Anorexia Nervosa/genetics , Diseases in Twins/genetics , Humans , Quality of Life , Risk Factors , Twins, Monozygotic/genetics
4.
J Psychosom Res ; 126: 109797, 2019 11.
Article in English | MEDLINE | ID: mdl-31536865

ABSTRACT

OBJECTIVES: Gastrointestinal problems are common in all eating disorders; however, the extent to which these problems predate the onset of eating disorders is not clear. We explored longitudinal associations between childhood gastrointestinal problems and adolescent disordered eating, and assessed whether observed associations are potentially causal or due to familial confounding factors. METHODS: Data from a population-based Swedish twin sample were used to investigate associations between parent- and self-reported protracted constipation and diarrhea in childhood and adolescence, and later disordered eating, measured by the Eating Disorders Inventory-2 (EDI). Linear regression models were used to investigate the associations. Possible familial confounding was explored by using a within-twin pair analysis. RESULTS: We found that those who reported a history of constipation at age 15 scored 5.55 and 5.04 points higher, respectively, on the EDI total score at age 15 and 18, compared with those without constipation. Those reporting a history of diarrhea at age 15 scored 5.15 points higher, and the group reporting both problems scored 9.52 points higher on the EDI total score at age 15 than those reporting no problems. We observed that the association between constipation and disordered eating was attenuated in the within-twin pair analysis, but remained positive. CONCLUSIONS: Gastrointestinal problems in childhood and adolescence are significantly associated with disordered eating. Associations were partly due to familial confounding, but might also be consistent with a causal interpretation. Clinicians should be aware of the increased risk of disordered eating when following children and adolescents who present with gastrointestinal problems.


Subject(s)
Constipation/complications , Diarrhea/complications , Feeding and Eating Disorders/physiopathology , Adolescent , Female , Humans , Longitudinal Studies , Male
5.
Cell ; 178(4): 850-866.e26, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398340

ABSTRACT

We performed a comprehensive assessment of rare inherited variation in autism spectrum disorder (ASD) by analyzing whole-genome sequences of 2,308 individuals from families with multiple affected children. We implicate 69 genes in ASD risk, including 24 passing genome-wide Bonferroni correction and 16 new ASD risk genes, most supported by rare inherited variants, a substantial extension of previous findings. Biological pathways enriched for genes harboring inherited variants represent cytoskeletal organization and ion transport, which are distinct from pathways implicated in previous studies. Nevertheless, the de novo and inherited genes contribute to a common protein-protein interaction network. We also identified structural variants (SVs) affecting non-coding regions, implicating recurrent deletions in the promoters of DLG2 and NR3C2. Loss of nr3c2 function in zebrafish disrupts sleep and social function, overlapping with human ASD-related phenotypes. These data support the utility of studying multiplex families in ASD and are available through the Hartwell Autism Research and Technology portal.


Subject(s)
Autism Spectrum Disorder/genetics , Genetic Predisposition to Disease/genetics , Pedigree , Protein Interaction Maps/genetics , Animals , Child , Databases, Genetic , Disease Models, Animal , Female , Gene Deletion , Guanylate Kinases/genetics , Humans , Inheritance Patterns/genetics , Machine Learning , Male , Nuclear Family , Promoter Regions, Genetic/genetics , Receptors, Mineralocorticoid/genetics , Risk Factors , Tumor Suppressor Proteins/genetics , Whole Genome Sequencing , Zebrafish/genetics
6.
Nat Genet ; 51(8): 1207-1214, 2019 08.
Article in English | MEDLINE | ID: mdl-31308545

ABSTRACT

Characterized primarily by a low body-mass index, anorexia nervosa is a complex and serious illness1, affecting 0.9-4% of women and 0.3% of men2-4, with twin-based heritability estimates of 50-60%5. Mortality rates are higher than those in other psychiatric disorders6, and outcomes are unacceptably poor7. Here we combine data from the Anorexia Nervosa Genetics Initiative (ANGI)8,9 and the Eating Disorders Working Group of the Psychiatric Genomics Consortium (PGC-ED) and conduct a genome-wide association study of 16,992 cases of anorexia nervosa and 55,525 controls, identifying eight significant loci. The genetic architecture of anorexia nervosa mirrors its clinical presentation, showing significant genetic correlations with psychiatric disorders, physical activity, and metabolic (including glycemic), lipid and anthropometric traits, independent of the effects of common variants associated with body-mass index. These results further encourage a reconceptualization of anorexia nervosa as a metabo-psychiatric disorder. Elucidating the metabolic component is a critical direction for future research, and paying attention to both psychiatric and metabolic components may be key to improving outcomes.


Subject(s)
Anorexia Nervosa/etiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Genomics/methods , Mental Disorders/complications , Metabolic Diseases/complications , Quantitative Trait Loci , Adult , Anorexia Nervosa/genetics , Anorexia Nervosa/pathology , Body Mass Index , Case-Control Studies , Female , Humans , Male , Mental Disorders/genetics , Metabolic Diseases/genetics , Phenotype , Prognosis
7.
Focus (Am Psychiatr Publ) ; 17(1): 66-72, 2019 Jan.
Article in English | MEDLINE | ID: mdl-32015716

ABSTRACT

(Gandal et al., "Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap" Science 09 Feb 2018:Vol. 359, Issue 6376, pp. 693-697 (DOI: 10.1126/science.aad6469). Reprinted with permission from AAAS).

8.
Contemp Clin Trials ; 74: 61-69, 2018 11.
Article in English | MEDLINE | ID: mdl-30287268

ABSTRACT

BACKGROUND: Genetic factors contribute to anorexia nervosa (AN); and the first genome-wide significant locus has been identified. We describe methods and procedures for the Anorexia Nervosa Genetics Initiative (ANGI), an international collaboration designed to rapidly recruit 13,000 individuals with AN and ancestrally matched controls. We present sample characteristics and the utility of an online eating disorder diagnostic questionnaire suitable for large-scale genetic and population research. METHODS: ANGI recruited from the United States (US), Australia/New Zealand (ANZ), Sweden (SE), and Denmark (DK). Recruitment was via national registers (SE, DK); treatment centers (US, ANZ, SE, DK); and social and traditional media (US, ANZ, SE). All cases had a lifetime AN diagnosis based on DSM-IV or ICD-10 criteria (excluding amenorrhea). Recruited controls had no lifetime history of disordered eating behaviors. To assess the positive and negative predictive validity of the online eating disorder questionnaire (ED100K-v1), 109 women also completed the Structured Clinical Interview for DSM-IV (SCID), Module H. RESULTS: Blood samples and clinical information were collected from 13,363 individuals with lifetime AN and from controls. Online diagnostic phenotyping was effective and efficient; the validity of the questionnaire was acceptable. CONCLUSIONS: Our multi-pronged recruitment approach was highly effective for rapid recruitment and can be used as a model for efforts by other groups. High online presence of individuals with AN rendered the Internet/social media a remarkably effective recruitment tool in some countries. ANGI has substantially augmented Psychiatric Genomics Consortium AN sample collection. ANGI is a registered clinical trial: clinicaltrials.govNCT01916538; https://clinicaltrials.gov/ct2/show/NCT01916538?cond=Anorexia+Nervosa&draw=1&rank=3.


Subject(s)
Anorexia Nervosa/diagnosis , Adolescent , Adult , Aged , Anorexia Nervosa/genetics , Australia , Case-Control Studies , Denmark , Feeding and Eating Disorders/diagnosis , Female , Humans , Internet , Middle Aged , New Zealand , Patient Selection , Reproducibility of Results , Surveys and Questionnaires , Sweden , United States , Young Adult
9.
Int J Eat Disord ; 51(7): 593-607, 2018 07.
Article in English | MEDLINE | ID: mdl-30194862

ABSTRACT

OBJECTIVE: We explored both within-method and between-method rigor and reproducibility in the field of eating disorders genetics. METHOD: We present critical evaluation and commentary on component methods of genetic research (family studies, twin studies, molecular genetic studies) and discuss both successful and unsuccessful efforts in the field. RESULTS: Eating disorders genetics has had a number of robust results that converge across component methodologies. Familial aggregation of eating disorders, twin-based heritability estimates of eating disorders, and genome-wide association studies (GWAS) all point toward a substantial role for genetics in eating disorders etiology and support the premise that genes do not act alone. Candidate gene and linkage studies have been less informative historically. DISCUSSION: The eating disorders field has entered the GWAS era with studies of anorexia nervosa. Continued growth of sample sizes is essential for rigorous discovery of actionable variation. Molecular genetic studies of bulimia nervosa, binge-eating disorder, and other eating disorders are virtually nonexistent and lag seriously behind other major psychiatric disorders. Expanded efforts are necessary to reveal the fundamental biology of eating disorders, inform clinical practice, and deliver new therapeutic targets.


Subject(s)
Anorexia Nervosa/genetics , Bulimia Nervosa/genetics , Feeding and Eating Disorders/genetics , Genetic Research , Reproducibility of Results , Bayes Theorem , Databases, Factual , Genetic Linkage , Genome-Wide Association Study , Humans
10.
Nature ; 560(7718): E30, 2018 08.
Article in English | MEDLINE | ID: mdl-29995847

ABSTRACT

Change history: In this Letter, the labels for splicing events A3SS and A5SS were swapped in column D of Supplementary Table 3a and b. This has been corrected online.

11.
Science ; 359(6376): 693-697, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29439242

ABSTRACT

The predisposition to neuropsychiatric disease involves a complex, polygenic, and pleiotropic genetic architecture. However, little is known about how genetic variants impart brain dysfunction or pathology. We used transcriptomic profiling as a quantitative readout of molecular brain-based phenotypes across five major psychiatric disorders-autism, schizophrenia, bipolar disorder, depression, and alcoholism-compared with matched controls. We identified patterns of shared and distinct gene-expression perturbations across these conditions. The degree of sharing of transcriptional dysregulation is related to polygenic (single-nucleotide polymorphism-based) overlap across disorders, suggesting a substantial causal genetic component. This comprehensive systems-level view of the neurobiological architecture of major neuropsychiatric illness demonstrates pathways of molecular convergence and specificity.


Subject(s)
Genetic Predisposition to Disease , Mental Disorders/genetics , Multifactorial Inheritance , Nervous System Diseases/genetics , Cerebral Cortex/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Polymorphism, Single Nucleotide , Transcription, Genetic
12.
Nature ; 540(7633): 423-427, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27919067

ABSTRACT

Autism spectrum disorder (ASD) involves substantial genetic contributions. These contributions are profoundly heterogeneous but may converge on common pathways that are not yet well understood. Here, through post-mortem genome-wide transcriptome analysis of the largest cohort of samples analysed so far, to our knowledge, we interrogate the noncoding transcriptome, alternative splicing, and upstream molecular regulators to broaden our understanding of molecular convergence in ASD. Our analysis reveals ASD-associated dysregulation of primate-specific long noncoding RNAs (lncRNAs), downregulation of the alternative splicing of activity-dependent neuron-specific exons, and attenuation of normal differences in gene expression between the frontal and temporal lobes. Our data suggest that SOX5, a transcription factor involved in neuron fate specification, contributes to this reduction in regional differences. We further demonstrate that a genetically defined subtype of ASD, chromosome 15q11.2-13.1 duplication syndrome (dup15q), shares the core transcriptomic signature observed in idiopathic ASD. Co-expression network analysis reveals that individuals with ASD show age-related changes in the trajectory of microglial and synaptic function over the first two decades, and suggests that genetic risk for ASD may influence changes in regional cortical gene expression. Our findings illustrate how diverse genetic perturbations can lead to phenotypic convergence at multiple biological levels in a complex neuropsychiatric disorder.


Subject(s)
Alternative Splicing/genetics , Autism Spectrum Disorder/genetics , Gene Expression Profiling , Gene Expression Regulation , Genome, Human/genetics , RNA, Long Noncoding/genetics , Animals , Autopsy , Case-Control Studies , Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , Exons/genetics , Frontal Lobe/metabolism , Humans , Intellectual Disability/genetics , Neurons/metabolism , Primates/genetics , SOXD Transcription Factors/metabolism , Species Specificity , Temporal Lobe/metabolism , Transcriptome/genetics
13.
Nat Neurosci ; 19(11): 1397-1407, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27786179

ABSTRACT

Hundreds of genetic loci increasing risk for neuropsychiatric disorders have recently been identified. This success, perhaps paradoxically, has posed challenges for therapeutic development, which are amplified by the highly polygenic and pleiotropic nature of these genetic contributions. Success requires understanding the biological impact of single genetic variants and predicting their effects within an individual. Comprehensive functional genomic annotation of risk loci provides a framework for interpretation of neurobiological impact, requiring experimental validation with in vivo or in vitro model systems. Systems-level, integrative pathway analyses are beginning to elucidate the additive, polygenic contributions of risk variants on specific cellular, molecular, developmental, or circuit-level processes. Although most neuropsychiatric disease modeling has focused on genes disrupted by rare, large-effect-size mutations, common smaller-effect-size variants may also provide solid therapeutic targets to inform precision medicine approaches. Here we enumerate the promise and challenges of a genomics-driven approach to uncovering neuropsychiatric disease mechanisms and facilitating therapeutic development.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Multifactorial Inheritance/genetics , Precision Medicine , Psychotherapeutic Processes , Animals , Humans , Mutation/genetics
14.
Am J Hum Genet ; 99(3): 540-554, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27569545

ABSTRACT

Rare mutations, including copy-number variants (CNVs), contribute significantly to autism spectrum disorder (ASD) risk. Although their importance has been established in families with only one affected child (simplex families), the contribution of both de novo and inherited CNVs to ASD in families with multiple affected individuals (multiplex families) is less well understood. We analyzed 1,532 families from the Autism Genetic Resource Exchange (AGRE) to assess the impact of de novo and rare CNVs on ASD risk in multiplex families. We observed a higher burden of large, rare CNVs, including inherited events, in individuals with ASD than in their unaffected siblings (odds ratio [OR] = 1.7), but the rate of de novo events was significantly lower than in simplex families. In previously characterized ASD risk loci, we identified 49 CNVs, comprising 24 inherited events, 19 de novo events, and 6 events of unknown inheritance, a significant enrichment in affected versus control individuals (OR = 3.3). In 21 of the 30 families (71%) in whom at least one affected sibling harbored an established ASD major risk CNV, including five families harboring inherited CNVs, the CNV was not shared by all affected siblings, indicating that other risk factors are contributing. We also identified a rare risk locus for ASD and language delay at chromosomal region 2q24 (implicating NR4A2) and another lower-penetrance locus involving inherited deletions and duplications of WWOX. The genetic architecture in multiplex families differs from that in simplex families and is complex, warranting more complete genetic characterization of larger multiplex ASD cohorts.


Subject(s)
Autism Spectrum Disorder/genetics , DNA Copy Number Variations/genetics , Genetic Predisposition to Disease/genetics , Chromosomes, Human, Pair 2/genetics , Cohort Studies , Databases, Genetic , Exons/genetics , Female , Gene Duplication/genetics , Genome-Wide Association Study , Humans , Language Development Disorders/genetics , Male , Odds Ratio , Oligonucleotide Array Sequence Analysis , Oxidoreductases/genetics , Penetrance , Promoter Regions, Genetic/genetics , Risk Factors , Sequence Deletion/genetics , Siblings , Tumor Suppressor Proteins/genetics , Untranslated Regions/genetics , WW Domain-Containing Oxidoreductase
15.
Nat Neurosci ; 16(9): 1228-1237, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23912948

ABSTRACT

Implicating particular genes in the generation of complex brain and behavior phenotypes requires multiple lines of evidence. The rarity of most high-impact genetic variants typically precludes the possibility of accruing statistical evidence that they are associated with a given trait. We found that the enrichment of a rare chromosome 22q11.22 deletion in a recently expanded Northern Finnish sub-isolate enabled the detection of association between TOP3B and both schizophrenia and cognitive impairment. Biochemical analysis of TOP3ß revealed that this topoisomerase was a component of cytosolic messenger ribonucleoproteins (mRNPs) and was catalytically active on RNA. The recruitment of TOP3ß to mRNPs was independent of RNA cis-elements and was coupled to the co-recruitment of FMRP, the disease gene product in fragile X mental retardation syndrome. Our results indicate a previously unknown role for TOP3ß in mRNA metabolism and suggest that it is involved in neurodevelopmental disorders.


Subject(s)
Abnormalities, Multiple/genetics , Cognition Disorders/genetics , DNA Topoisomerases, Type I/genetics , DiGeorge Syndrome/genetics , Schizophrenia/genetics , Sequence Deletion/genetics , Adolescent , Adult , Aged , Chromosome Deletion , Chromosomes, Human, Pair 22/genetics , Cognition Disorders/epidemiology , Cohort Studies , Family Health , Female , Finland/epidemiology , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Gene Expression Profiling , Genetic Association Studies , Genotype , HEK293 Cells , Health Surveys , Humans , Male , Middle Aged , Models, Molecular , Proteins/genetics , Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Schizophrenia/epidemiology , Young Adult
16.
Nat Genet ; 45(6): 670-5, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23603763

ABSTRACT

Primary sclerosing cholangitis (PSC) is a severe liver disease of unknown etiology leading to fibrotic destruction of the bile ducts and ultimately to the need for liver transplantation. We compared 3,789 PSC cases of European ancestry to 25,079 population controls across 130,422 SNPs genotyped using the Immunochip. We identified 12 genome-wide significant associations outside the human leukocyte antigen (HLA) complex, 9 of which were new, increasing the number of known PSC risk loci to 16. Despite comorbidity with inflammatory bowel disease (IBD) in 72% of the cases, 6 of the 12 loci showed significantly stronger association with PSC than with IBD, suggesting overlapping yet distinct genetic architectures for these two diseases. We incorporated association statistics from 7 diseases clinically occurring with PSC in the analysis and found suggestive evidence for 33 additional pleiotropic PSC risk loci. Together with network analyses, these findings add to the genetic risk map of PSC and expand on the relationship between PSC and other immune-mediated diseases.


Subject(s)
Cholangitis, Sclerosing/genetics , Case-Control Studies , Cholangitis, Sclerosing/immunology , Gene Frequency , Genetic Loci/immunology , Genetic Pleiotropy , Genome-Wide Association Study , Genotyping Techniques , Humans , Linkage Disequilibrium , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Risk Factors
17.
BMJ Open ; 1(1): e000087, 2011 Aug 27.
Article in English | MEDLINE | ID: mdl-22021758

ABSTRACT

Objectives Depression is a worldwide leading cause of morbidity and disability. Genetic studies have recently begun to elucidate its molecular aetiology. The authors investigated candidate genes of monoamine neurotransmission and early environmental risk factors for depressiveness in the genetically isolated population-based Northern Finland Birth Cohort 1966 (12 058 live births). Design The authors ascertained and subdivided the study sample (n=5225) based on measures of early development and of social environment, and examined candidate genes of monoamine neurotransmission, many of which have shown prior evidence of a gene-environment interaction for affective disorders, namely SLC6A4, TPH2, COMT, MAOA and the dopamine receptor genes DRD1-DRD5. Results and conclusion The authors observed no major genetic effects of the analysed variants on depressiveness. However, when measures of early development and of social environment were considered, some evidence of interaction was observed. Allelic variants of COMT interacted with high early developmental risk (p=0.005 for rs2239393 and p=0.02 for rs4680) so that the association with depression was detected only in individuals at high developmental risk group (p=0.0046 and ß=0.056 for rs5993883-rs2239393-rs4680 risk haplotype CGG including Val158), particularly in males (p=0.0053 and ß=0.083 for the haplotype CGG). Rs4274224 from DRD2 interacted with gender (p=0.017) showing a significant association with depressiveness in males (p=0.0006 and ß=0.0023; p=0.00005 and ß=0.069 for rs4648318-rs4274224 haplotype GG). The results support the role of genes of monoamine neurotransmission in the aetiology of depression conditional on environmental risk and sex, but not direct major effects of monoaminergic genes in this unselected population.

19.
Am J Hum Genet ; 86(2): 285-91, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20159113

ABSTRACT

Genetic risk for multiple sclerosis (MS) is thought to involve both common and rare risk alleles. Recent GWAS and subsequent meta-analysis have established the critical role of the HLA locus and identified new common variants associated to MS. These variants have small odds ratios (ORs) and explain only a fraction of the genetic risk. To expose potentially rare, high-impact alleles, we conducted a GWAS of 68 distantly related cases and 136 controls from a high-risk internal isolate of Finland with increased prevalence and familial occurrence of MS. The top 27 loci with p < 10(-4) were tested in 711 cases and 1029 controls from Finland, and the top two findings were validated in 3859 cases and 9110 controls from more heterogeneous populations. SNP (rs744166) within the STAT3 gene was associated to MS (p = 2.75 x 10(-10), OR 0.87, confidence interval 0.83-0.91). The protective haplotype for MS in STAT3 is a risk allele for Crohn disease, implying that STAT3 represents a shared risk locus for at least two autoimmune diseases. This study also demonstrates the potential of special isolated populations in search for variants contributing to complex traits.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide/genetics , STAT3 Transcription Factor/genetics , Alleles , Base Pairing/genetics , Case-Control Studies , Genetics, Population , Haplotypes/genetics , Humans , Linkage Disequilibrium/genetics , Reproducibility of Results
20.
Proc Natl Acad Sci U S A ; 106(13): 5264-9, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19237575

ABSTRACT

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system associated with demyelination and axonal loss. A whole genome association scan suggested that allelic variants in the CD58 gene region, encoding the costimulatory molecule LFA-3, are associated with risk of developing MS. We now report additional genetic evidence, as well as resequencing and fine mapping of the CD58 locus in patients with MS and control subjects. These efforts identify a CD58 variant that provides further evidence of association with MS (P = 1.1 x 10(-6), OR 0.82) and the single protective effect within the CD58 locus is captured by the rs2300747(G) allele. This protective rs2300747(G) allele is associated with a dose-dependent increase in CD58 mRNA expression in lymphoblastic cell lines (P = 1.1 x 10(-10)) and in peripheral blood mononuclear cells from MS subjects (P = 0.0037). This protective effect of enhanced CD58 expression on circulating mononuclear cells in patients with MS is supported by finding that CD58 mRNA expression is higher in MS subjects during clinical remission. Functional investigations suggest a potential mechanism whereby increases in CD58 expression, mediated by the protective allele, up-regulate the expression of transcription factor FoxP3 through engagement of the CD58 receptor, CD2, leading to the enhanced function of CD4(+)CD25(high) regulatory T cells that are defective in subjects with MS.


Subject(s)
CD58 Antigens/genetics , Multiple Sclerosis/genetics , RNA, Messenger/analysis , Alleles , CD2 Antigens , Case-Control Studies , Chromosome Mapping , Forkhead Transcription Factors , Humans , Leukocytes, Mononuclear , Remission Induction , Sequence Analysis , T-Lymphocytes, Regulatory/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...