Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 146(4): 1403-1419, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-36152315

RESUMEN

Genome-wide association studies have identified 10q24.32 as a robust schizophrenia risk locus. Here we identify a regulatory variant (rs10786700) that disrupts binding of transcription factors at 10q24.32. We independently confirmed the association between rs10786700 and schizophrenia in a large Chinese cohort (n = 11 547) and uncovered the biological mechanism underlying this association. We found that rs10786700 resides in a super-enhancer element that exhibits dynamic activity change during the development process and that the risk allele (C) of rs10786700 conferred significant lower enhancer activity through enhancing binding affinity to repressor element-1 silencing transcription factor (REST). CRISPR-Cas9-mediated genome editing identified SUFU as a potential target gene by which rs10786700 might exert its risk effect on schizophrenia, as deletion of rs10786700 downregulated SUFU expression. We further investigated the role of Sufu in neurodevelopment and found that Sufu knockdown inhibited proliferation of neural stem cells and neurogenesis, affected molecular pathways (including neurodevelopment-related pathways, PI3K-Akt and ECM-receptor interaction signalling pathways) associated with schizophrenia and altered the density of dendritic spines. These results reveal that the functional risk single nucleotide polymorphism rs10786700 at 10q24.32 interacts with REST synergistically to regulate expression of SUFU, a novel schizophrenia risk gene which is involved in schizophrenia pathogenesis by affecting neurodevelopment and spine morphogenesis.


Asunto(s)
Esquizofrenia , Humanos , Esquizofrenia/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Fosfatidilinositol 3-Quinasas/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Transcripción/genética
2.
Hum Genet ; 142(6): 809-818, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37085628

RESUMEN

Immune dysregulation has been consistently reported in psychiatric disorders, however, the causes and mechanisms underlying immune dysregulation in psychiatric disorders remain largely unclear. Here we conduct a Mendelian randomization study by integrating plasma proteome and GWASs of schizophrenia, bipolar disorder and depression. The primate-specific immune-related protein BTN3A3 showed the most significant associations with all three psychiatric disorders. In addition, other immune-related proteins, including AIF1, FOXO3, IRF3, CFHR4, IGLON5, FKBP2, and PI3, also showed significant associations with psychiatric disorders. Our study showed that a proportion of psychiatric risk variants may contribute to disease risk by regulating immune-related plasma proteins, providing direct evidence that connect the genetic risk of psychiatric disorders to immune system.


Asunto(s)
Trastorno Bipolar , Trastornos Mentales , Animales , Proteoma/genética , Proteoma/metabolismo , Análisis de la Aleatorización Mendeliana , Trastornos Mentales/genética , Trastorno Bipolar/genética , Proteínas Sanguíneas , Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple
3.
Brain ; 145(2): 770-786, 2022 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-34581804

RESUMEN

Genome-wide association studies have shown that genetic variants at 2q33.1 are strongly associated with schizophrenia. However, potential causal variants in this locus and their roles in schizophrenia remain unknown. Here, we identified two functional variants (rs796364 and rs281759) that disrupt CTCF, RAD21 and FOXP2 binding at 2q33.1. We systematically investigated the regulatory mechanisms of these two variants with serial experiments, including reporter gene assays and electrophoretic mobility shift assay. Intriguingly, these two single nucleotide polymorphisms physically interacted with TYW5 and showed the most significant associations with TYW5 expression in human brain. Consistently, CRISPR-Cas9-mediated genome editing confirmed the regulatory effect of the two single nucleotide polymorphisms on TYW5 expression. Additionally, expression analysis indicated that TYW5 was significantly upregulated in brains of schizophrenia cases compared with controls, suggesting that rs796364 and rs281759 might confer schizophrenia risk by modulating TYW5 expression. We over-expressed TYW5 in mouse neural stem cells and rat primary neurons to mimic its upregulation in schizophrenia and found significant alterations in the proliferation and differentiation of neural stem cells, as well as dendritic spine density following TYW5 overexpression, indicating its important roles in neurodevelopment and spine morphogenesis. Furthermore, we independently confirmed the association between rs796364 and schizophrenia in a Chinese cohort of 8202 subjects. Finally, transcriptome analysis revealed that TYW5 affected schizophrenia-associated pathways. These lines of evidence consistently revealed that rs796364 and rs281759 might contribute to schizophrenia risk by regulating the expression of TYW5, a gene whose expression dysregulation affects two important schizophrenia pathophysiological processes (i.e. neurodevelopment and dendritic spine formation).


Asunto(s)
Estudio de Asociación del Genoma Completo , Oxigenasas de Función Mixta/genética , Esquizofrenia , Animales , Pueblo Asiatico/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Ratones , Polimorfismo de Nucleótido Simple/genética , Ratas , Esquizofrenia/genética
4.
Brain ; 145(7): 2569-2585, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35094059

RESUMEN

Recent genome-wide association studies have reported multiple schizophrenia risk loci, yet the functional variants and their roles in schizophrenia remain to be characterized. Here we identify a functional single nucleotide polymorphism (rs2270363: G>A) at the schizophrenia risk locus 16p13.3. rs2270363 lies in the E-box element of the promoter of NMRAL1 and disrupts binding of the basic helix-loop-helix leucine zipper family proteins, including USF1, MAX and MXI1. We validated the regulatory effects of rs2270363 using reporter gene assays and electrophoretic mobility shift assay. Besides, expression quantitative trait loci analysis showed that the risk allele (A) of rs2270363 was significantly associated with elevated NMRAL1 expression in the human brain. Transcription factors knockdown and CRISPR-Cas9-mediated editing further confirmed the regulatory effects of the genomic region containing rs2270363 on NMRAL1. Intriguingly, NMRAL1 was significantly downregulated in the brain of schizophrenia patients compared with healthy subjects, and knockdown of Nmral1 expression affected proliferation and differentiation of mouse neural stem cells, as well as genes and pathways associated with brain development and synaptic transmission. Of note, Nmral1 knockdown resulted in significant decrease of dendritic spine density, revealing the potential pathophysiological mechanisms of NMRAL1 in schizophrenia. Finally, we independently confirmed the association between rs2270363 and schizophrenia in the Chinese population and found that the risk allele of rs2270363 was the same in European and Chinese populations. These lines of evidence suggest that rs2270363 may confer schizophrenia risk by regulating NMRAL1, a gene whose expression dysregulation might be involved in the pathogenesis of schizophrenia by affecting neurodevelopment and synaptic plasticity.


Asunto(s)
Esquizofrenia , Factores de Transcripción , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Factores de Transcripción/genética
5.
BMC Med ; 20(1): 68, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35168626

RESUMEN

BACKGROUND: Genome-wide association studies (GWASs) have identified multiple risk loci for Parkinson's disease (PD). However, identifying the functional (or potential causal) variants in the reported risk loci and elucidating their roles in PD pathogenesis remain major challenges. To identify the potential causal (or functional) variants in the reported PD risk loci and to elucidate their regulatory mechanisms, we report a functional genomics study of PD. METHODS: We first integrated chromatin immunoprecipitation sequencing (ChIP-Seq) (from neuronal cells and human brain tissues) data and GWAS-identified single-nucleotide polymorphisms (SNPs) in PD risk loci. We then conducted a series of experiments and analyses to validate the regulatory effects of these (i.e., functional) SNPs, including reporter gene assays, allele-specific expression (ASE), transcription factor (TF) knockdown, CRISPR-Cas9-mediated genome editing, and expression quantitative trait loci (eQTL) analysis. RESULTS: We identified 44 SNPs (from 11 risk loci) affecting the binding of 12 TFs and we validated the regulatory effects of 15 TF binding-disrupting SNPs. In addition, we also identified the potential target genes regulated by these TF binding-disrupting SNPs through eQTL analysis. Finally, we showed that 4 eQTL genes of these TF binding-disrupting SNPs were dysregulated in PD cases compared with controls. CONCLUSION: Our study systematically reveals the gene regulatory mechanisms of PD risk variants (including widespread disruption of CTCF binding), generates the landscape of potential PD causal variants, and pinpoints promising candidate genes for further functional characterization and drug development.


Asunto(s)
Estudio de Asociación del Genoma Completo , Enfermedad de Parkinson , Predisposición Genética a la Enfermedad/genética , Genómica , Humanos , Enfermedad de Parkinson/genética , Polimorfismo de Nucleótido Simple/genética
6.
Mov Disord ; 37(12): 2451-2456, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36177513

RESUMEN

BACKGROUND: Large-scale genome-wide association studies (GWASs) have reported multiple risk variants for Parkinson's disease (PD). However, little is known about how these reported risk variants confer risk of PD. OBJECTIVE: To nominate genes whose genetically regulated expression in dopaminergic neurons may have a causal role in PD. METHODS: We conducted a two-sample Mendelian randomization (MR) study by integrating large-scale genome-wide associations and expression quantitative trait loci (eQTL) data from dopaminergic neurons. RESULTS: MR analysis nominated 10 risk genes whose genetically regulated expression in dopaminergic neurons may have a causal role in PD. These MR significant genes include FAM200B, NDUFAF2, NUP42, SH3GL2, STX1B, CCDC189, KAT8, PRSS36, VAMP4, and ZSWIM7. CONCLUSIONS: We report the first MR study of PD by using dopaminergic neuron-specific eQTL and nominate novel risk genes for PD. Further functional characterization of the nominated risk genes will provide mechanistic insights into PD pathogenesis and potential therapeutic targets. © 2022 International Parkinson and Movement Disorder Society.


Asunto(s)
Estudio de Asociación del Genoma Completo , Enfermedad de Parkinson , Humanos , Sitios de Carácter Cuantitativo/genética , Análisis de la Aleatorización Mendeliana , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Neuronas Dopaminérgicas/metabolismo , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple
7.
Mol Psychiatry ; 26(11): 6896-6911, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33931730

RESUMEN

Genome-wide association studies (GWASs) have revealed that genetic variants at the 22q13.2 risk locus were robustly associated with schizophrenia. However, the causal variants at this risk locus and their roles in schizophrenia remain elusive. Here we identify the risk missense variant rs1801311 (located in the 1st exon of NDUFA6 gene) as likely causal for schizophrenia at 22q13.2 by disrupting binding of YY1, TAF1, and POLR2A. We systematically elucidated the regulatory mechanisms of rs1801311 and validated the regulatory effect of this missense variant. Intriguingly, rs1801311 physically interacted with NAGA (encodes the alpha-N-acetylgalactosaminidase, which is mainly involved in regulating metabolisms of glycoproteins and glycolipids in lysosome) and showed the most significant association with NAGA expression in the human brain, with the risk allele (G) associated with higher NAGA expression. Consistent with eQTL analysis, expression analysis showed that NAGA was significantly upregulated in brains of schizophrenia cases compared with controls, further supporting that rs1801311 may confer schizophrenia risk by regulating NAGA expression. Of note, we found that NAGA regulates important neurodevelopmental processes, including proliferation and differentiation of neural stem cells. Transcriptome analysis corroborated that NAGA regulates pathways associated with neuronal differentiation. Finally, we independently confirmed the association between rs1801311 and schizophrenia in a large Chinese cohort. Our study elucidates the regulatory mechanisms of the missense schizophrenia risk variant rs1801311 and provides mechanistic links between risk variant and schizophrenia etiology. In addition, this study also revealed the novel role of coding variants in gene regulation and schizophrenia risk, i.e., genetic variant in coding region of a specific gene may confer disease risk through regulating distal genes (act as regulatory variant for distal genes).


Asunto(s)
Esquizofrenia , Alelos , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Mutación Missense/genética , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia/genética , Factor de Transcripción YY1/genética , alfa-N-Acetilgalactosaminidasa/genética , alfa-N-Acetilgalactosaminidasa/metabolismo
8.
BMC Med ; 19(1): 177, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34380480

RESUMEN

BACKGROUND: Over 200 schizophrenia risk loci have been identified by genome-wide association studies (GWASs). However, the majority of risk loci were identified in populations of European ancestry (EUR), potentially missing important biological insights. It is important to perform 5 GWASs in non-European populations. METHODS: To identify novel schizophrenia risk loci, we conducted a GWAS in Han Chinese population (3493 cases and 4709 controls). We then performed a large-scale meta-analysis (a total of 143,438 subjects) through combining our results with previous GWASs conducted in EAS and EUR. In addition, we also carried out comprehensive post-GWAS analysis, including heritability partitioning, enrichment of schizophrenia associations in tissues and cell types, trancscriptome-wide association study (TWAS), expression quantitative trait loci (eQTL) and differential expression analysis. RESULTS: We identified two new schizophrenia risk loci, including associations in SHISA9 (rs7192086, P = 4.92 × 10-08) and PES1 (rs57016637, P = 2.33 × 10-11) in Han Chinese population. A fixed-effect meta-analysis (a total of 143,438 subjects) with summary statistics from EAS and EUR identifies 15 novel genome-wide significant risk loci. Heritability partitioning with linkage disequilibrium score regression (LDSC) reveals a significant enrichment of schizophrenia heritability in conserved genomic regions, promoters, and enhancers. Tissue and cell-type enrichment analyses show that schizophrenia associations are significantly enriched in human brain tissues and several types of neurons, including cerebellum neurons, telencephalon inhibitory, and excitatory neurons. Polygenic risk score profiling reveals that GWAS summary statistics from trans-ancestry meta-analysis (EAS + EUR) improves prediction performance in predicting the case/control status of our sample. Finally, transcriptome-wide association study (TWAS) identifies risk genes whose cis-regulated expression change may have a role in schizophrenia. CONCLUSIONS: Our study identifies 17 novel schizophrenia risk loci and highlights the importance and necessity of conducting genetic study in different populations. These findings not only provide new insights into genetic etiology of schizophrenia, but also facilitate to delineate the pathophysiology of schizophrenia and develop new therapeutic targets.


Asunto(s)
Estudio de Asociación del Genoma Completo , Esquizofrenia , Estudios de Casos y Controles , Predisposición Genética a la Enfermedad , Humanos , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Proteínas de Unión al ARN , Esquizofrenia/genética , Transcriptoma
9.
Mol Psychiatry ; 25(9): 1926-1945, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32214206

RESUMEN

Major depressive disorder (MDD) is one of the most prevalent psychiatric disorders and a leading cause of disability worldwide. Though recent genome-wide association studies (GWAS) have identified multiple risk variants for MDD, how these variants confer MDD risk remains largely unknown. Here we systematically characterize the regulatory mechanism of MDD risk variants using a functional genomics approach. By integrating chromatin immunoprecipitation sequencing (ChIP-Seq) (from human brain tissues or neuronal cells) and position weight matrix (PWM) data, we identified 34 MDD risk SNPs that disrupt the binding of 15 transcription factors (TFs). We verified the regulatory effect of the TF binding-disrupting SNPs with reporter gene assays, allelic-specific expression analysis, and CRISPR-Cas9-mediated genome editing. Expression quantitative trait loci (eQTL) analysis identified the target genes that might be regulated by these regulatory risk SNPs. Finally, we found that NEGR1 (regulated by the TF binding-disrupting MDD risk SNP rs3101339) was dysregulated in the brains of MDD cases compared with controls, implying that rs3101339 may confer MDD risk by affecting NEGR1 expression. Our findings reveal how genetic variants contribute to MDD risk by affecting TF binding and gene regulation. More importantly, our study identifies the potential MDD causal variants and their target genes, thus providing pivotal candidates for future mechanistic study and drug development.


Asunto(s)
Trastorno Depresivo Mayor , Estudio de Asociación del Genoma Completo , Trastorno Depresivo Mayor/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética
10.
Am J Med Genet B Neuropsychiatr Genet ; 186(1): 28-39, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522098

RESUMEN

Schizophrenia is a severe mental disease characterized with positive symptoms, negative symptoms, and cognitive impairments. Although recent genome-wide association studies (GWASs) have identified over 145 risk loci for schizophrenia, pinpointing the causal variants and genes at the reported loci and elucidating their roles in schizophrenia remain major challenges. Here we identify a functional single-nucleotide polymorphism (SNP; rs213237) in ZNF323 promoter by using functional fine-mapping. We found that allelic differences at rs213237 affected the ZNF323 promoter activity significantly. Consistently, expression quantitative trait loci (eQTL) analysis showed that rs213237 was significantly associated with ZNF323 expression in diverse human brain tissues, suggesting that rs213237 may contribute to schizophrenia risk through regulating ZNF323 expression. Interestingly, we found that ZNF323 protein was localized in the nucleus and knockdown of ZNF323 in macaque neural stem cells (mNSCs) significantly impaired proliferation and survival of mNSCs. We further showed that stable knockdown of ZNF323 in SH-SY5Y cells resulted in significant decrease of the tyrosine hydroxylase (TH) protein expression. Finally, transcriptome analysis revealed that ZNF323 may regulate pivotal schizophrenia risk genes (including VIPR2 and NPY) and schizophrenia-associated pathways (including PI3K-AKT and NOTCH signaling pathways), suggesting that ZNF323 may be a major regulator of schizophrenia risk genes. Our study reveals how a genetic variant in ZNF323 promoter contributes to schizophrenia risk through regulating ZNF323 expression. More importantly, our findings demonstrate that ZNF323 may have a pivotal role in schizophrenia pathogenesis through regulating schizophrenia risk genes and schizophrenia-associated biological processes (including neurodevelopment, PI3K-AKT, and NOTCH signaling pathways).


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Estudio de Asociación del Genoma Completo , Células-Madre Neurales/patología , Neuroblastoma/patología , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Esquizofrenia/patología , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Humanos , Macaca , Células-Madre Neurales/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Fenotipo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Factores de Transcripción/genética , Células Tumorales Cultivadas
11.
Hum Genet ; 139(10): 1285-1297, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32385526

RESUMEN

During the past decade, genetic studies of schizophrenia have become one of the most exciting and fast-moving areas. Hundreds of genes implicated in schizophrenia have been identified by genetic, epigenetic, and gene expression studies. However, how to systematically and efficiently use these published data to pinpoint the causal genes becomes a major challenge in schizophrenia research. Here, we release an updated version of a comprehensive database for schizophrenia research, SZDB2.0 ( www.szdb.org ), which accompanies significant data expansion and feature improvements, as well as functionality optimization. Compared with the first version (SZDB), the current database has the following updates: (1) We added the newly published genome-wide association study (GWAS) of schizophrenia from CLOZUK + PGC, which is the largest GWAS for schizophrenia; (2) We included a polygenic risk score calculator; (3) In the refined "Gene" module of SZDB2.0, we collated genetic, gene expression, methylation, and integrative results of all available schizophrenia studies; (4) In the "CNV (copy number variation)" module, we collated the results of all 77 CNV publications about schizophrenia; (5) We also updated other data, including gene expression quantitative trait loci (eQTL), transcript QTL, methylation QTL, and protein-protein interaction data, based on the information from the latest literatures. We optimized the query interface of SZDB2.0 for a better visualization and data retrieval. The updated SZDB2.0 will advance the research of schizophrenia.


Asunto(s)
Bases de Datos Genéticas , Epigénesis Genética , Predisposición Genética a la Enfermedad , Herencia Multifactorial , Sitios de Carácter Cuantitativo , Esquizofrenia/genética , Variaciones en el Número de Copia de ADN , Metilación de ADN , Ontología de Genes , Estudio de Asociación del Genoma Completo , Humanos , Almacenamiento y Recuperación de la Información/métodos , Polimorfismo de Nucleótido Simple , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Mapeo de Interacción de Proteínas , Riesgo , Esquizofrenia/diagnóstico , Esquizofrenia/fisiopatología
14.
Hereditas ; 155: 16, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29308060

RESUMEN

BACKGROUND: Recent large-scale genome-wide association studies (GWAS) have showed that the neuronal calcium signaling has pivotal roles in schizophrenia (SCZ) in populations of European of ancestry. However, it is not known if calcium signaling pathway genes are also associated with SCZ in Han Chinese population. METHODS: Here we investigated the association between genetic variants in three calcium signaling pathway genes (CACNB2, CACNA1C and CACNA1I) and SCZ in 1615 SCZ cases and 1597 controls. RESULTS: A single nucleotide polymorphism (SNP) (rs4522708) in CACNA1I is significantly associated with SCZ in our Chinese sample (ORA allele = 1.19, corrected P = 0.042), suggesting that CACNA1I may also be a risk gene for SCZ in Chinese population. Of note, the risk allele (A allele) of SNP rs4522708 is same in European and Chinese populations. Meta-analysis of Chinese and European samples further strengthened the association of rs4522708 with SCZ (ORA allele = 1.074, P = 6.26 × 10-11). Expression analysis showed that CACNA1I was significantly up-regulated in hippocampus of SCZ cases compared with controls, implying that dysregulation of CACNA1I may have a role in schizophrenia pathogenesis. CONCLUSIONS: Our study suggests that CACNA1I is a risk gene for SCZ in Chinese population and provides further evidence that supports the potential role of neuronal calcium signaling in schizophrenia.


Asunto(s)
Canales de Calcio Tipo T/genética , Predisposición Genética a la Enfermedad , Esquizofrenia/genética , Alelos , Pueblo Asiatico/genética , Señalización del Calcio , Estudios de Casos y Controles , China , Genotipo , Humanos , Polimorfismo de Nucleótido Simple
16.
Hum Genet ; 135(4): 377-392, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26875095

RESUMEN

Greatly expanded brain volume is one of the most characteristic traits that distinguish humans from other primates. Recent studies have revealed genes responsible for the dramatically enlarged human brain size (i.e., the microcephaly genes), and it has been well documented that many microcephaly genes have undergone accelerated evolution along the human lineage. In addition to being far larger than other primates, human brain volume is also highly variable in general populations. However, the genetic basis underlying human brain volume variation remains elusive and it is not known whether genes regulating human brain volume variation also have experienced positive selection. We have previously shown that genetic variants (near the IL3 gene) on 5q33 were significantly associated with brain volume in Chinese population. Here, we provide further evidence that support the significant association of genetic variants on 5q33 with brain volume. Bioinformatic analyses suggested that rs31480 is likely to be the causal variant among the studied SNPs. Molecular evolutionary analyses suggested that IL3 might have undergone positive selection in primates and humans. Neutrality tests further revealed signatures of positive selection of IL3 in Han Chinese and Europeans. Finally, extended haplotype homozygosity (EHH) and relative EHH analyses showed that the C allele of SNP rs31480 might have experienced recent positive selection in Han Chinese. Our results suggest that IL3 is an important genetic regulator for human brain volume variation and implied that IL3 might have experienced weak or modest positive selection in the evolutionary history of humans, which may be due to its contribution to human brain volume.


Asunto(s)
Adaptación Fisiológica , Encéfalo/anatomía & histología , Evolución Molecular , Interleucina-3/genética , Secuencia de Aminoácidos , Animales , China , Humanos , Interleucina-3/química , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
17.
Br J Psychiatry ; 208(2): 128-37, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26338991

RESUMEN

BACKGROUND: Bipolar disorder is a highly heritable polygenic disorder. Recent enrichment analyses suggest that there may be true risk variants for bipolar disorder in the expression quantitative trait loci (eQTL) in the brain. AIMS: We sought to assess the impact of eQTL variants on bipolar disorder risk by combining data from both bipolar disorder genome-wide association studies (GWAS) and brain eQTL. METHOD: To detect single nucleotide polymorphisms (SNPs) that influence expression levels of genes associated with bipolar disorder, we jointly analysed data from a bipolar disorder GWAS (7481 cases and 9250 controls) and a genome-wide brain (cortical) eQTL (193 healthy controls) using a Bayesian statistical method, with independent follow-up replications. The identified risk SNP was then further tested for association with hippocampal volume (n = 5775) and cognitive performance (n = 342) among healthy individuals. RESULTS: Integrative analysis revealed a significant association between a brain eQTL rs6088662 on chromosome 20q11.22 and bipolar disorder (log Bayes factor = 5.48; bipolar disorder P = 5.85 × 10(-5)). Follow-up studies across multiple independent samples confirmed the association of the risk SNP (rs6088662) with gene expression and bipolar disorder susceptibility (P = 3.54 × 10(-8)). Further exploratory analysis revealed that rs6088662 is also associated with hippocampal volume and cognitive performance in healthy individuals. CONCLUSIONS: Our findings suggest that 20q11.22 is likely a risk region for bipolar disorder; they also highlight the informative value of integrating functional annotation of genetic variants for gene expression in advancing our understanding of the biological basis underlying complex disorders, such as bipolar disorder.


Asunto(s)
Trastorno Bipolar/genética , Cognición , Hipocampo/patología , Polimorfismo de Nucleótido Simple , Anciano , Teorema de Bayes , Estudios de Casos y Controles , Cromosomas Humanos Par 22 , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Modelos Logísticos
18.
Am J Med Genet B Neuropsychiatr Genet ; 171B(2): 160-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26437209

RESUMEN

Previous studies have suggested that genetic variants for schizophrenia susceptibility might contribute to structural brain volume variations in schizophrenia patients, including total brain volume, hippocampal volume, and amygdalar volume. However, whether these schizophrenia susceptibility variants are associated with macroscopic structural brain volume (i.e., intracranial volume, total brain volume, and hippocampal volume) in healthy subjects is still unclear. In this study, we investigated the associations between 47 schizophrenia susceptibility variants (from 25 well-characterized schizophrenia susceptibility genes) and cranial volume variation in a healthy Chinese sample (N = 1,013). We also extracted the association between these 47 schizophrenia risk variants and the macroscopic structural brain volume (intracranial volume, total brain volume and hippocampal volume) in a large healthy sample of European ancestry (ENIGMA sample, N = 5,775). We identified several single-nucleotide polymorphisms (SNPs) nominally associated with intracranial volume, total brain volume, and hippocampal volume at P < 0.05 (uncorrected). However, after Bonferroni corrections for multiple testing, no SNP showed significant association. Hence, our results do not support previous observations that schizophrenia susceptibility variants are associated with brain structure (e.g., hippocampal volume) in healthy individuals, and indicate that single schizophrenia risk variant may not contribute significantly to macroscopic brain structure (e.g., intracranial volume or hippocampal volume) variation in healthy subjects.


Asunto(s)
Encéfalo/patología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Salud , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia/genética , Adulto , Pueblo Asiatico/genética , Femenino , Humanos , Masculino , Tamaño de los Órganos , Adulto Joven
19.
J Neurosci ; 34(30): 10072-7, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25057208

RESUMEN

In mammals, formation of the auditory sensory organ (the organ of Corti) is restricted to a specialized area of the cochlea. However, the molecular mechanisms limiting sensory formation to this discrete region in the ventral cochlear duct are not well understood, nor is it known whether other regions of the cochlea have the competence to form the organ of Corti. Here we identify LMO4, a LIM-domain-only nuclear protein, as a negative regulator of sensory organ formation in the cochlea. Inactivation of Lmo4 in mice leads to an ectopic organ of Corti (eOC) located in the lateral cochlea. The eOC retains the features of the native organ, including inner and outer hair cells, supporting cells, and other nonsensory specialized cell types. However, the eOC shows an orientation opposite to the native organ, such that the eOC appears as a mirror-image duplication to the native organ of Corti. These data demonstrate a novel sensory competent region in the lateral cochlear duct that is regulated by LMO4 and may be amenable to therapeutic manipulation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas con Dominio LIM/genética , Órgano Espiral/crecimiento & desarrollo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Cóclea/crecimiento & desarrollo , Femenino , Técnicas de Sustitución del Gen , Proteínas con Dominio LIM/antagonistas & inhibidores , Proteínas con Dominio LIM/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Órganos de los Sentidos/crecimiento & desarrollo
20.
Hum Mol Genet ; 22(18): 3609-23, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23666531

RESUMEN

HDR syndrome (also known as Barakat syndrome) is a developmental disorder characterized by hypoparathyroidism, sensorineural deafness and renal disease. Although genetic mapping and subsequent functional studies indicate that GATA3 haplo-insufficiency causes human HDR syndrome, the role of Gata3 in sensorineural deafness and auditory system development is largely unknown. In this study, we show that Gata3 is continuously expressed in the developing mouse inner ear. Conditional knockout of Gata3 in the developing inner ear disrupts the morphogenesis of mouse inner ear, resulting in a disorganized and shortened cochlear duct with significant fewer hair cells and supporting cells. Loss of Gata3 function leads to the failure in the specification of prosensory domain and subsequently, to increased cell death in the cochlear duct. Moreover, though the initial generation of cochleovestibular ganglion (CVG) cells is not affected in Gata3-null mice, spiral ganglion neurons (SGNs) are nearly depleted due to apoptosis. Our results demonstrate the essential role of Gata3 in specifying the prosensory domain in the cochlea and in regulating the survival of SGNs, thus identifying a molecular mechanism underlying human HDR syndrome.


Asunto(s)
Conducto Coclear/embriología , Oído Interno/embriología , Oído Interno/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Células Receptoras Sensoriales/fisiología , Animales , Apoptosis , Conducto Coclear/citología , Conducto Coclear/inervación , Modelos Animales de Enfermedad , Oído Interno/inervación , Regulación del Desarrollo de la Expresión Génica , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Hipoparatiroidismo/genética , Hipoparatiroidismo/metabolismo , Ratones , Ratones Noqueados , Nefrosis/genética , Nefrosis/metabolismo , Ganglio Espiral de la Cóclea/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA