Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.550
Filter
1.
Front Public Health ; 12: 1327315, 2024.
Article in English | MEDLINE | ID: mdl-38827616

ABSTRACT

Background: Substantial research evidence supports the correlation between mental disorders and sepsis. Nevertheless, the causal connection between a particular psychological disorder and sepsis remains unclear. Methods: For investigating the causal relationships between mental disorders and sepsis, genetic variants correlated with mental disorders, including anorexia nervosa (AN), attention-deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD), bipolar disorder (BD), major depressive disorder (MDD), obsessive-compulsive disorder (OCD), panic disorder (PD), posttraumatic stress disorder (PTSD), schizophrenia (SCZ), and tourette syndrome (TS), were all extracted from the Psychiatric Genomics Consortium (PGC). The causal estimates and direction between these mental disorders and sepsis were evaluated employing a two-sample bidirectional MR strategy. The inverse variance weighted (IVW) method was the primary approach utilized. Various sensitivity analyses were performed to confirm the validity of the causal effect. Meta-analysis, multivariable MR, and mediation MR were conducted to ensure the credibility and depth of this research. Results: The presence of AN was in relation to a greater likelihood of sepsis (OR 1.08, 95% CI 1.02-1.14; p = 0.013). A meta-analysis including validation cohorts supported this observation (OR 1.06, 95% CI 1.02-1.09). None of the investigated mental disorders appeared to be impacted when sepsis was set as the exposure factor. Even after adjusting for confounding factors, AN remained statistically significant (OR 1.08, 95% CI 1.02-1.15; p = 0.013). Mediation analysis indicated N-formylmethionine levels (with a mediated proportion of 7.47%), cystatin D levels (2.97%), ketogluconate Metabolism (17.41%) and N10-formyl-tetrahydrofolate biosynthesis (20.06%) might serve as mediators in the pathogenesis of AN-sepsis. Conclusion: At the gene prediction level, two-sample bidirectional MR analysis revealed that mental disorder AN had a causal association with an increased likelihood of sepsis. In addition, N-formylmethionine levels, cystatin D levels, ketogluconate metabolism and N10-formyl-tetrahydrofolate biosynthesis may function as potential mediators in the pathophysiology of AN-sepsis. Our research may contribute to the investigation of novel therapeutic strategies for mental illness and sepsis.


Subject(s)
Mendelian Randomization Analysis , Mental Disorders , Sepsis , Humans , Mental Disorders/genetics , Female
2.
Transl Psychiatry ; 14(1): 236, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830862

ABSTRACT

Recent genetic studies have found common genomic risk variants among psychiatric disorders, strongly suggesting the overlaps in their molecular and cellular mechanism. Our research group identified the variant in ASTN2 as one of the candidate risk factors across these psychiatric disorders by whole-genome copy number variation analysis. However, the alterations in the human neuronal cells resulting from ASTN2 variants identified in patients remain unknown. To address this, we used patient-derived and genome-edited iPS cells with ASTN2 deletion; cells were further differentiated into neuronal cells. A comprehensive gene expression analysis using genome-edited iPS cells with variants on both alleles revealed that the expression level of ZNF558, a gene specifically expressed in human forebrain neural progenitor cells, was greatly reduced in ASTN2-deleted neuronal cells. Furthermore, the expression of the mitophagy-related gene SPATA18, which is repressed by ZNF558, and mitophagy activity were increased in ASTN2-deleted neuronal cells. These phenotypes were also detected in neuronal cells differentiated from patient-derived iPS cells with heterozygous ASTN2 deletion. Our results suggest that ASTN2 deletion is related to the common pathogenic mechanism of psychiatric disorders by regulating mitophagy via ZNF558.


Subject(s)
Induced Pluripotent Stem Cells , Mental Disorders , Neurons , Humans , Induced Pluripotent Stem Cells/metabolism , Mental Disorders/genetics , Neurons/metabolism , Neural Stem Cells/metabolism , Cell Differentiation/genetics , DNA Copy Number Variations , Gene Deletion , Transcription Factors/genetics
3.
Biochem J ; 481(10): 615-642, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38722301

ABSTRACT

Early-life adversities, whether prenatal or postnatal exposure, have been linked to adverse mental health outcomes later in life increasing the risk of several psychiatric disorders. Research on its neurobiological consequences demonstrated an association between exposure to adversities and persistent alterations in the structure, function, and connectivity of the brain. Consistent evidence supports the idea that regulation of gene expression through epigenetic mechanisms are involved in embedding the impact of early-life experiences in the genome and mediate between social environments and later behavioral phenotypes. In addition, studies from rodent models and humans suggest that these experiences and the acquired risk factors can be transmitted through epigenetic mechanisms to offspring and the following generations potentially contributing to a cycle of disease or disease risk. However, one of the important aspects of epigenetic mechanisms, unlike genetic sequences that are fixed and unchangeable, is that although the epigenetic markings are long-lasting, they are nevertheless potentially reversible. In this review, we summarize our current understanding of the epigenetic mechanisms involved in the mental health consequences derived from early-life exposure to malnutrition, maltreatment and poverty, adversities with huge and pervasive impact on mental health. We also discuss the evidence about transgenerational epigenetic inheritance in mammals and experimental data suggesting that suitable social and pharmacological interventions could reverse adverse epigenetic modifications induced by early-life negative social experiences. In this regard, these studies must be accompanied by efforts to determine the causes that promote these adversities and that result in health inequity in the population.


Subject(s)
Epigenesis, Genetic , Mental Disorders , Humans , Animals , Mental Disorders/genetics , Mental Disorders/etiology , Mental Health , Prenatal Exposure Delayed Effects/genetics , Pregnancy , Female , Adverse Childhood Experiences , DNA Methylation
4.
Transl Psychiatry ; 14(1): 221, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811559

ABSTRACT

Substance use disorder (SUD) is a global health problem with a significant impact on individuals and society. The presentation of SUD is diverse, involving various substances, ages at onset, comorbid conditions, and disease trajectories. Current treatments for SUD struggle to address this heterogeneity, resulting in high relapse rates. SUD often co-occurs with other psychiatric and mental health-related conditions that contribute to the heterogeneity of the disorder and predispose to adverse disease trajectories. Family and genetic studies highlight the role of genetic and environmental factors in the course of SUD, and point to a shared genetic liability between SUDs and comorbid psychopathology. In this study, we aimed to disentangle SUD heterogeneity using a deeply phenotyped SUD cohort and polygenic scores (PGSs) for psychiatric disorders and related traits. We explored associations between PGSs and various SUD-related phenotypes, as well as PGS-environment interactions using information on lifetime emotional, physical, and/or sexual abuse. Our results identify clusters of individuals who exhibit differences in their phenotypic profile and reveal different patterns of associations between SUD-related phenotypes and the genetic liability for mental health-related traits, which may help explain part of the heterogeneity observed in SUD. In our SUD sample, we found associations linking the genetic liability for attention-deficit hyperactivity disorder (ADHD) with lower educational attainment, the genetic liability for post-traumatic stress disorder (PTSD) with higher rates of unemployment, the genetic liability for educational attainment with lower rates of criminal records and unemployment, and the genetic liability for well-being with lower rates of outpatient treatments and fewer problems related to family and social relationships. We also found evidence of PGS-environment interactions showing that genetic liability for suicide attempts worsened the psychiatric status in SUD individuals with a history of emotional physical and/or sexual abuse. Collectively, these data contribute to a better understanding of the role of genetic liability for mental health-related conditions and adverse life experiences in SUD heterogeneity.


Subject(s)
Multifactorial Inheritance , Phenotype , Substance-Related Disorders , Humans , Substance-Related Disorders/genetics , Substance-Related Disorders/epidemiology , Male , Female , Adult , Genetic Predisposition to Disease , Middle Aged , Genome-Wide Association Study , Gene-Environment Interaction , Young Adult , Comorbidity , Mental Disorders/genetics , Mental Disorders/epidemiology
5.
Cells ; 13(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38786025

ABSTRACT

Stress is a common denominator of complex disorders and the FK-506 binding protein (FKBP)51 plays a central role in stress. Hence, it is not surprising that multiple studies imply the involvement of the FKBP51 protein and/or its coding gene, FKBP5, in complex disorders. This review summarizes such reports concentrating on three disorder clusters-neuropsychiatric, cancer, and type 2 diabetes mellitus (T2DM). We also attempt to point to potential mechanisms suggested to mediate the effect of FKBP5/FKBP51 on these disorders. Neuropsychiatric diseases considered in this paper include (i) Huntington's disease for which increased autophagic cellular clearance mechanisms related to decreased FKBP51 protein levels or activity is discussed, Alzheimer's disease for which increased FKBP51 activity has been shown to induce Tau phosphorylation and aggregation, and Parkinson's disease in the context of which FKBP12 is mentioned; and (ii) mental disorders, for which significant association with the single nucleotide polymorphism (SNP) rs1360780 of FKBP5 intron 7 along with decreased DNA methylation were revealed. Since cancer is a large group of diseases that can start in almost any organ or tissue of the body, FKBP51's role depends on the tissue type and differences among pathways expressed in those tumors. The FKBP51-heat-shock protein-(Hsp)90-p23 super-chaperone complex might function as an oncogene or as a tumor suppressor by downregulating the serine/threonine protein kinase (AKt) pathway. In T2DM, two potential pathways for the involvement of FKBP51 are highlighted as affecting the pathogenesis of the disease-the peroxisome proliferator-activated receptor-γ (PPARγ) and AKt.


Subject(s)
Diabetes Mellitus, Type 2 , Mental Disorders , Neoplasms , Tacrolimus Binding Proteins , Humans , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Tacrolimus Binding Proteins/metabolism , Tacrolimus Binding Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Mental Disorders/genetics , Mental Disorders/metabolism , Animals
6.
Brain Behav ; 14(5): e3532, 2024 May.
Article in English | MEDLINE | ID: mdl-38779749

ABSTRACT

BACKGROUND: Neurocognitive and psychiatric disorders have been proved that they can comorbid more often with idiopathic normal pressure hydrocephalus (iNPH) than general population. However, the potential causal association between these disorders and iNPH has not been assessed. Thus, our study aims to investigate the causal relationship between them based on a bidirectional Mendelian randomization (MR) analysis. METHODS: Random effects of the inverse variance weighted (IVW) method were conducted to obtain the causal association among the neurocognitive disorders, psychiatric disorders, and iNPH. Genome-wide association studies (GWAS) of 12 neurocognitive and psychiatric disorders were downloaded via the OpenGWAS database, GWAS Catalog, and Psychiatric Genomics Consortium, whereas GWAS data of iNPH were obtained from the FinnGen consortium round 9 release, with 767 cases and 375,610 controls of European ancestry. We also conducted the sensitivity analysis in these significant causal inferences using weighted median model, Cochrane's Q test, MR-Egger regression, MR Pleiotropy Residual Sum and Outlier detect and the leave-one-out analysis. RESULTS: For most of the neurocognitive and psychiatric disorders, no causal association was established between them and iNPH. We have found that iNPH (odds ratio [OR] = 1.030, 95% confidence interval [CI]: 1.011-1.048, p = .001) is associated with increased risk for schizophrenia, which failed in validation of sensitivity analysis. Notably, genetically predicted Parkinson's disease (PD) is associated with increased risk of iNPH (OR = 1.256, 95% CI: 1.045-1.511, p = .015). CONCLUSION: Our study has revealed the potential causal effect in which PD associated with an increased risk of iNPH. Further study is warranted to investigate the association between PD and iNPH and the potential underlying mechanism.


Subject(s)
Genome-Wide Association Study , Hydrocephalus, Normal Pressure , Mendelian Randomization Analysis , Mental Disorders , Humans , Hydrocephalus, Normal Pressure/genetics , Hydrocephalus, Normal Pressure/epidemiology , Mental Disorders/genetics , Mental Disorders/epidemiology , Neurocognitive Disorders/genetics , Neurocognitive Disorders/epidemiology
7.
Sci Transl Med ; 16(749): eadh9974, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38781321

ABSTRACT

Many psychiatric disorders exhibit sex differences, but the underlying mechanisms remain poorly understood. We analyzed transcriptomics data from 2160 postmortem adult prefrontal cortex brain samples from the PsychENCODE consortium in a sex-stratified study design. We compared transcriptomics data of postmortem brain samples from patients with schizophrenia (SCZ), bipolar disorder (BD), and autism spectrum disorder (ASD) with transcriptomics data of postmortem control brains from individuals without a known history of psychiatric disease. We found that brain samples from females with SCZ, BD, and ASD showed a higher burden of transcriptomic dysfunction than did brain samples from males with these disorders. This observation was supported by the larger number of differentially expressed genes (DEGs) and a greater magnitude of gene expression changes observed in female versus male brain specimens. In addition, female patient brain samples showed greater overall connectivity dysfunction, defined by a higher proportion of gene coexpression modules with connectivity changes and higher connectivity burden, indicating a greater degree of gene coexpression variability. We identified several gene coexpression modules enriched in sex-biased DEGs and identified genes from a genome-wide association study that were involved in immune and synaptic functions across different brain cell types. We found a number of genes as hubs within these modules, including those encoding SCN2A, FGF14, and C3. Our results suggest that in the context of psychiatric diseases, males and females exhibit different degrees of transcriptomic dysfunction and implicate immune and synaptic-related pathways in these sex differences.


Subject(s)
Autopsy , Brain , Mental Disorders , Sex Characteristics , Transcriptome , Humans , Female , Male , Transcriptome/genetics , Brain/metabolism , Brain/pathology , Mental Disorders/genetics , Mental Disorders/pathology , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Gene Expression Profiling , Genome-Wide Association Study , Adult , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Gene Regulatory Networks , Middle Aged
8.
PLoS One ; 19(5): e0247212, 2024.
Article in English | MEDLINE | ID: mdl-38753848

ABSTRACT

We investigated the functional classes of genomic regions containing SNPS contributing most to the SNP-heritability of important psychiatric and neurological disorders and behavioral traits, as determined from recent genome-wide association studies. We employed linkage-disequilibrium score regression with several brain-specific genomic annotations not previously utilized. The classes of genomic annotations conferring substantial SNP-heritability for the psychiatric disorders and behavioral traits differed systematically from the classes associated with neurological disorders, and both differed from the classes enriched for height, a biometric trait used here as a control outgroup. The SNPs implicated in these psychiatric disorders and behavioral traits were highly enriched in CTCF binding sites, in conserved regions likely to be enhancers, and in brain-specific promoters, regulatory sites likely to affect responses to experience. The SNPs relevant for neurological disorders were highly enriched in constitutive coding regions and splice regulatory sites.


Subject(s)
Genome-Wide Association Study , Mental Disorders , Nervous System Diseases , Polymorphism, Single Nucleotide , Humans , Mental Disorders/genetics , Nervous System Diseases/genetics , Linkage Disequilibrium , Genetic Predisposition to Disease , Promoter Regions, Genetic
9.
Biol Sex Differ ; 15(1): 42, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750598

ABSTRACT

BACKGROUND: Sex differences exist in the prevalence and clinical manifestation of several mental disorders, suggesting that sex-specific brain phenotypes may play key roles. Previous research used machine learning models to classify sex from imaging data of the whole brain and studied the association of class probabilities with mental health, potentially overlooking regional specific characteristics. METHODS: We here investigated if a regionally constrained model of brain volumetric imaging data may provide estimates that are more sensitive to mental health than whole brain-based estimates. Given its known role in emotional processing and mood disorders, we focused on the limbic system. Using two different cohorts of healthy subjects, the Human Connectome Project and the Queensland Twin IMaging, we investigated sex differences and heritability of brain volumes of limbic structures compared to non-limbic structures, and subsequently applied regionally constrained machine learning models trained solely on limbic or non-limbic features. To investigate the biological underpinnings of such models, we assessed the heritability of the obtained sex class probability estimates, and we investigated the association with major depression diagnosis in an independent clinical sample. All analyses were performed both with and without controlling for estimated total intracranial volume (eTIV). RESULTS: Limbic structures show greater sex differences and are more heritable compared to non-limbic structures in both analyses, with and without eTIV control. Consequently, machine learning models performed well at classifying sex based solely on limbic structures and achieved performance as high as those on non-limbic or whole brain data, despite the much smaller number of features in the limbic system. The resulting class probabilities were heritable, suggesting potentially meaningful underlying biological information. Applied to an independent population with major depressive disorder, we found that depression is associated with male-female class probabilities, with largest effects obtained using the limbic model. This association was significant for models not controlling for eTIV whereas in those controlling for eTIV the associations did not pass significance correction. CONCLUSIONS: Overall, our results highlight the potential utility of regionally constrained models of brain sex to better understand the link between sex differences in the brain and mental disorders.


Psychiatric disorders have different prevalence between sexes, with women being twice as likely to develop depression and anxiety across the lifespan. Previous studies have investigated sex differences in brain structure that might contribute to this prevalence but have mostly focused on a single-structure level, potentially overlooking the interplay between brain regions. Sex differences in structures responsible for emotional regulation (limbic system), affected in many psychiatric disorders, have been previously reported. Here, we apply a machine learning model to obtain an estimate of brain sex for each participant based on the volumes of multiple brain regions. Particularly, we compared the estimates obtained with a model based solely on limbic structures with those obtained with a non-limbic model (entire brain except limbic structures) and a whole brain model. To investigate the genetic determinants of the models, we assessed the heritability of the estimates between identical twins and fraternal twins. The estimates of all our models were heritable, suggesting a genetic component contributing to brain sex. Finally, to investigate the association with mental health, we compared brain sex estimates in healthy subjects and in a depressed population. We found an association between depression and brain sex in females for the limbic model, but not for the non-limbic model. No effect was found in males. Overall, our results highlight the potential utility of machine learning models of brain sex based on relevant structures to better understand the link between sex differences in the brain and mental disorders.


Subject(s)
Limbic System , Mental Disorders , Phenotype , Sex Characteristics , Humans , Limbic System/diagnostic imaging , Female , Male , Mental Disorders/genetics , Mental Disorders/diagnostic imaging , Adult , Machine Learning , Depressive Disorder, Major/genetics , Depressive Disorder, Major/diagnostic imaging , Young Adult , Middle Aged
10.
Twin Res Hum Genet ; 27(2): 85-96, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38699821

ABSTRACT

TwinsMX registry is a national research initiative in Mexico that aims to understand the complex interplay between genetics and environment in shaping physical and mental health traits among the country's population. With a multidisciplinary approach, TwinsMX aims to advance our knowledge of the genetic and environmental mechanisms underlying ethnic variations in complex traits and diseases, including behavioral, psychometric, anthropometric, metabolic, cardiovascular and mental disorders. With information gathered from over 2800 twins, this article updates the prevalence of several complex traits; and describes the advances and novel ideas we have implemented such as magnetic resonance imaging. The future expansion of the TwinsMX registry will enhance our comprehension of the intricate interplay between genetics and environment in shaping health and disease in the Mexican population. Overall, this report describes the progress in the building of a solid database that will allow the study of complex traits in the Mexican population, valuable not only for our consortium, but also for the worldwide scientific community, by providing new insights of understudied genetically admixed populations.


Subject(s)
Gene-Environment Interaction , Registries , Humans , Mexico/epidemiology , Male , Female , Adult , Diseases in Twins/genetics , Diseases in Twins/epidemiology , Middle Aged , Twins, Monozygotic/genetics , Twins, Dizygotic/genetics , Mental Disorders/genetics , Mental Disorders/epidemiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/epidemiology
11.
Nat Commun ; 15(1): 3803, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778015

ABSTRACT

Human endogenous retroviruses (HERVs) are repetitive elements previously implicated in major psychiatric conditions, but their role in aetiology remains unclear. Here, we perform specialised transcriptome-wide association studies that consider HERV expression quantified to precise genomic locations, using RNA sequencing and genetic data from 792 post-mortem brain samples. In Europeans, we identify 1238 HERVs with expression regulated in cis, of which 26 represent expression signals associated with psychiatric disorders, with ten being conditionally independent from neighbouring expression signals. Of these, five are additionally significant in fine-mapping analyses and thus are considered high confidence risk HERVs. These include two HERV expression signatures specific to schizophrenia risk, one shared between schizophrenia and bipolar disorder, and one specific to major depressive disorder. No robust signatures are identified for autism spectrum conditions or attention deficit hyperactivity disorder in Europeans, or for any psychiatric trait in other ancestries, although this is likely a result of relatively limited statistical power. Ultimately, our study highlights extensive HERV expression and regulation in the adult cortex, including in association with psychiatric disorder risk, therefore providing a rationale for exploring neurological HERV expression in complex neuropsychiatric traits.


Subject(s)
Bipolar Disorder , Depressive Disorder, Major , Endogenous Retroviruses , Genome-Wide Association Study , Schizophrenia , Transcriptome , Humans , Endogenous Retroviruses/genetics , Schizophrenia/genetics , Schizophrenia/virology , Bipolar Disorder/genetics , Risk Factors , Depressive Disorder, Major/genetics , Depressive Disorder, Major/virology , Mental Disorders/genetics , Brain/metabolism , Brain/virology , Female , Male , Genetic Predisposition to Disease , Attention Deficit Disorder with Hyperactivity/genetics , Adult
12.
Science ; 384(6698): eadh7688, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781356

ABSTRACT

RNA splicing is highly prevalent in the brain and has strong links to neuropsychiatric disorders; yet, the role of cell type-specific splicing and transcript-isoform diversity during human brain development has not been systematically investigated. In this work, we leveraged single-molecule long-read sequencing to deeply profile the full-length transcriptome of the germinal zone and cortical plate regions of the developing human neocortex at tissue and single-cell resolution. We identified 214,516 distinct isoforms, of which 72.6% were novel (not previously annotated in Gencode version 33), and uncovered a substantial contribution of transcript-isoform diversity-regulated by RNA binding proteins-in defining cellular identity in the developing neocortex. We leveraged this comprehensive isoform-centric gene annotation to reprioritize thousands of rare de novo risk variants and elucidate genetic risk mechanisms for neuropsychiatric disorders.


Subject(s)
Mental Disorders , Neocortex , Neurogenesis , Protein Isoforms , RNA Splicing , Single-Cell Analysis , Transcriptome , Humans , Alternative Splicing , Genetic Predisposition to Disease , Mental Disorders/genetics , Molecular Sequence Annotation , Neocortex/metabolism , Neocortex/embryology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Neurogenesis/genetics
13.
Science ; 384(6698): eadi5199, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781369

ABSTRACT

Single-cell genomics is a powerful tool for studying heterogeneous tissues such as the brain. Yet little is understood about how genetic variants influence cell-level gene expression. Addressing this, we uniformly processed single-nuclei, multiomics datasets into a resource comprising >2.8 million nuclei from the prefrontal cortex across 388 individuals. For 28 cell types, we assessed population-level variation in expression and chromatin across gene families and drug targets. We identified >550,000 cell type-specific regulatory elements and >1.4 million single-cell expression quantitative trait loci, which we used to build cell-type regulatory and cell-to-cell communication networks. These networks manifest cellular changes in aging and neuropsychiatric disorders. We further constructed an integrative model accurately imputing single-cell expression and simulating perturbations; the model prioritized ~250 disease-risk genes and drug targets with associated cell types.


Subject(s)
Brain , Gene Regulatory Networks , Mental Disorders , Single-Cell Analysis , Humans , Aging/genetics , Brain/metabolism , Cell Communication/genetics , Chromatin/metabolism , Chromatin/genetics , Genomics , Mental Disorders/genetics , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Quantitative Trait Loci
14.
Science ; 384(6698): eadh0829, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781368

ABSTRACT

Neuropsychiatric genome-wide association studies (GWASs), including those for autism spectrum disorder and schizophrenia, show strong enrichment for regulatory elements in the developing brain. However, prioritizing risk genes and mechanisms is challenging without a unified regulatory atlas. Across 672 diverse developing human brains, we identified 15,752 genes harboring gene, isoform, and/or splicing quantitative trait loci, mapping 3739 to cellular contexts. Gene expression heritability drops during development, likely reflecting both increasing cellular heterogeneity and the intrinsic properties of neuronal maturation. Isoform-level regulation, particularly in the second trimester, mediated the largest proportion of GWAS heritability. Through colocalization, we prioritized mechanisms for about 60% of GWAS loci across five disorders, exceeding adult brain findings. Finally, we contextualized results within gene and isoform coexpression networks, revealing the comprehensive landscape of transcriptome regulation in development and disease.


Subject(s)
Alternative Splicing , Brain , Gene Expression Regulation, Developmental , Mental Disorders , Humans , Atlases as Topic , Autism Spectrum Disorder/genetics , Brain/metabolism , Brain/growth & development , Brain/embryology , Gene Regulatory Networks , Genome-Wide Association Study , Protein Isoforms/genetics , Protein Isoforms/metabolism , Quantitative Trait Loci , Schizophrenia/genetics , Transcriptome , Mental Disorders/genetics
15.
Science ; 384(6698): 858-859, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781384

ABSTRACT

A cell-by-cell look at neuropsychiatric diseases.


Subject(s)
Brain , Mental Disorders , Humans , Brain/growth & development , Brain/metabolism , Mental Disorders/genetics , Neurons/metabolism , Single-Cell Analysis
16.
Sci Adv ; 10(21): eadj4452, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781344

ABSTRACT

Most genetic variants associated with psychiatric disorders are located in noncoding regions of the genome. To investigate their functional implications, we integrate epigenetic data from the PsychENCODE Consortium and other published sources to construct a comprehensive atlas of candidate brain cis-regulatory elements. Using deep learning, we model these elements' sequence syntax and predict how binding sites for lineage-specific transcription factors contribute to cell type-specific gene regulation in various types of glia and neurons. The elements' evolutionary history suggests that new regulatory information in the brain emerges primarily via smaller sequence mutations within conserved mammalian elements rather than entirely new human- or primate-specific sequences. However, primate-specific candidate elements, particularly those active during fetal brain development and in excitatory neurons and astrocytes, are implicated in the heritability of brain-related human traits. Additionally, we introduce PsychSCREEN, a web-based platform offering interactive visualization of PsychENCODE-generated genetic and epigenetic data from diverse brain cell types in individuals with psychiatric disorders and healthy controls.


Subject(s)
Brain , Epigenesis, Genetic , Regulatory Sequences, Nucleic Acid , Humans , Brain/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Evolution, Molecular , Mental Disorders/genetics , Regulatory Elements, Transcriptional/genetics , Neurons/metabolism , Gene Expression Regulation , Transcription Factors/genetics , Transcription Factors/metabolism
17.
BMC Psychiatry ; 24(1): 304, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654235

ABSTRACT

BACKGROUND: Previous studies have reported associations between obstructive sleep apnea (OSA) and several mental disorders. However, further research is required to determine whether these associations are causal. Therefore, we evaluated the bidirectional causality between the genetic liability for OSA and nine mental disorders by using Mendelian randomization (MR). METHOD: We performed two-sample bidirectional MR of genetic variants for OSA and nine mental disorders. Summary statistics on OSA and the nine mental disorders were extracted from the FinnGen study and the Psychiatric Genomics Consortium. The primary analytical approach for estimating causal effects was the inverse-variance weighted (IVW), with the weighted median and MR Egger as complementary methods. The MR Egger intercept test, Cochran's Q test, Rucker's Q test, and the MR pleiotropy residual sum and outlier (MR-PRESSO) test were used for sensitivity analyses. RESULT: MR analyses showed that genetic liability for major depressive disorder (MDD) was associated with an increased risk of OSA (odds ratio [OR] per unit increase in the risk of MDD, 1.29; 95% CI, 1.11-1.49; P < 0.001). In addition, genetic liability for OSA may be associated with an increased risk of attention-deficit/hyperactivity disorder (ADHD) (OR = 1.26; 95% CI, 1.02-1.56; p = 0.032). There was no evidence that OSA is associated with other mental disorders. CONCLUSION: Our study indicated that genetic liability for MDD is associated with an increased risk of OSA without a bidirectional relationship. Additionally, there was suggestive evidence that genetic liability for OSA may have a causal effect on ADHD. These findings have implications for prevention and intervention strategies targeting OSA and ADHD. Further research is needed to investigate the biological mechanisms underlying our findings and the relationship between OSA and other mental disorders.


Subject(s)
Depressive Disorder, Major , Mendelian Randomization Analysis , Sleep Apnea, Obstructive , Humans , Sleep Apnea, Obstructive/genetics , Depressive Disorder, Major/genetics , Depressive Disorder, Major/epidemiology , Attention Deficit Disorder with Hyperactivity/genetics , Mental Disorders/genetics , Mental Disorders/epidemiology , Genetic Predisposition to Disease/genetics
18.
J Transl Med ; 22(1): 387, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664746

ABSTRACT

BACKGROUND: Integrating quantitative trait loci (QTL) data related to molecular phenotypes with genome-wide association study (GWAS) data is an important post-GWAS strategic approach employed to identify disease-associated molecular features. Various types of molecular phenotypes have been investigated in neuropsychiatric disorders. However, these findings pertaining to distinct molecular features are often independent of each other, posing challenges for having an overview of the mapped genes. METHODS: In this study, we comprehensively summarized published analyses focusing on four types of risk-related molecular features (gene expression, splicing transcriptome, protein abundance, and DNA methylation) across five common neuropsychiatric disorders. Subsequently, we conducted supplementary analyses with the latest GWAS dataset and corresponding deficient molecular phenotypes using Functional Summary-based Imputation (FUSION) and summary data-based Mendelian randomization (SMR). Based on the curated and supplemented results, novel reliable genes and their functions were explored. RESULTS: Our findings revealed that eQTL exhibited superior ability in prioritizing risk genes compared to the other QTL, followed by sQTL. Approximately half of the genes associated with splicing transcriptome, protein abundance, and DNA methylation were successfully replicated by eQTL-associated genes across all five disorders. Furthermore, we identified 436 novel reliable genes, which enriched in pathways related with neurotransmitter transportation such as synaptic, dendrite, vesicles, axon along with correlations with other neuropsychiatric disorders. Finally, we identified ten multiple molecular involved regulation patterns (MMRP), which may provide valuable insights into understanding the contribution of molecular regulation network targeting these disease-associated genes. CONCLUSIONS: The analyses prioritized novel and reliable gene sets related with five molecular features based on published and supplementary results for five common neuropsychiatric disorders, which were missed in the original GWAS analysis. Besides, the involved MMRP behind these genes could be given priority for further investigation to elucidate the pathogenic molecular mechanisms underlying neuropsychiatric disorders in future studies.


Subject(s)
DNA Methylation , Genetic Predisposition to Disease , Genome-Wide Association Study , Mental Disorders , Phenotype , Quantitative Trait Loci , Humans , Quantitative Trait Loci/genetics , Mental Disorders/genetics , DNA Methylation/genetics , Mendelian Randomization Analysis , Transcriptome/genetics
20.
Heart Lung ; 66: 86-93, 2024.
Article in English | MEDLINE | ID: mdl-38593678

ABSTRACT

BACKGROUND: Previous observational studies have suggested associations between Coronary Heart Disease (CHD) and Mental Health Disorders (MHD). However, the causal nature of these relationships has remained elusive. OBJECTIVE: The purpose of this study is to elucidate the causal relationships between eight distinct types of CHD and six types of MHD using Mendelian randomization (MR) analysis. METHODS: The MR analysis employed a suite of methods including inverse variance-weighted (IVW), MR-Egger, weighted mode, weighted median, and simple mode techniques. To assess heterogeneity, IVW and MR-Egger tests were utilized. MR-Egger regression also served to investigate potential pleiotropy. The stability of IVW results was verified by leave-one-out sensitivity analysis. RESULTS: We analyzed data from over 2,473,005 CHD and 803,801 MHD patients, informed by instrumental variables from large-scale genomic studies on European populations. The analysis revealed a causal increase in the risk of Major Depressive Disorder and Mania associated with Coronary Artery Disease and Myocardial Infarction. Heart Failure was found to causally increase the risk for Bipolar Disorder and Schizophrenia. Atrial Fibrillation and Ischemic Heart Diseases were positively linked to Generalized Anxiety Disorder and Mania, respectively. There was no significant evidence of an association between Hypertensive Heart Disease, Hypertrophic Cardiomyopathy, Pulmonary Heart Disease, and MHD. Reverse MR analysis indicated that MHD do not serve as risk factors for CHD. CONCLUSIONS: The findings suggest that specific types of CHD may act as risk factors for certain MHDs. Consequently, incorporating psychological assessments into the management of patients with CHD could be advantageous.


Subject(s)
Coronary Disease , Mendelian Randomization Analysis , Mental Disorders , Humans , Mendelian Randomization Analysis/methods , Coronary Disease/psychology , Coronary Disease/epidemiology , Coronary Disease/genetics , Coronary Disease/complications , Risk Factors , Mental Disorders/epidemiology , Mental Disorders/genetics , Male , Female
SELECTION OF CITATIONS
SEARCH DETAIL
...