Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Publication year range
1.
Anal Chem ; 96(11): 4430-4436, 2024 03 19.
Article in English | MEDLINE | ID: mdl-38447029

ABSTRACT

Traditional single-molecule fluorescence in situ hybridization (smFISH) methods for RNA detection often face sensitivity challenges due to the low fluorescence intensity of the probe. Also, short-lived autofluorescence complicates obtaining clear signals from tissue sections. In response, we have developed an smFISH probe using highly grafted lanthanide complexes to address both concentration quenching and autofluorescence background. Our approach involves an oligo PCR incorporating azide-dUTP, enabling conjugation with lanthanide complexes. This method has proven to be stable, convenient, and cost-effective. Notably, for the mRNA detection in SKBR3 cells, the lanthanide probe group exhibited 2.5 times higher luminescence intensity and detected 3 times more signal points in cells compared with the Cy3 group. Furthermore, we successfully applied the probe to image HER2 mRNA molecules in breast cancer FFPE tissue sections, achieving a 2.7-fold improvement in sensitivity compared to Cy3-based probes. These results emphasize the potential of time-resolved smFISH as a highly sensitive method for nucleic acid detection, free of background fluorescence interference.


Subject(s)
Lanthanoid Series Elements , In Situ Hybridization, Fluorescence/methods , RNA/analysis , RNA, Messenger/genetics , Diagnostic Imaging
2.
Brain ; 146(4): 1403-1419, 2023 04 19.
Article in English | MEDLINE | ID: mdl-36152315

ABSTRACT

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.


Subject(s)
Schizophrenia , Humans , Schizophrenia/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Phosphatidylinositol 3-Kinases/genetics , Polymorphism, Single Nucleotide/genetics , Transcription Factors/genetics
3.
Hum Genet ; 142(6): 809-818, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37085628

ABSTRACT

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.


Subject(s)
Bipolar Disorder , Mental Disorders , Animals , Proteome/genetics , Proteome/metabolism , Mendelian Randomization Analysis , Mental Disorders/genetics , Bipolar Disorder/genetics , Blood Proteins , Genome-Wide Association Study , Polymorphism, Single Nucleotide
4.
Psychol Med ; : 1-11, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37092861

ABSTRACT

BACKGROUND: To identify risk genes whose expression are regulated by the reported risk variants and to explore the potential regulatory mechanism in schizophrenia (SCZ). METHODS: We systematically integrated three independent brain expression quantitative traits (eQTLs) (CommonMind, GTEx, and BrainSeq Phase 2, a total of 1039 individuals) and GWAS data (56 418 cases and 78 818 controls), with the use of transcriptome-wide association study (TWAS). Diffusion magnetic resonance imaging was utilized to quantify the integrity of white matter bundles and determine whether polygenic risk of novel genes linked to brain structure was present in patients with first-episode antipsychotic SCZ. RESULTS: TWAS showed that eight risk genes (CORO7, DDAH2, DDHD2, ELAC2, GLT8D1, PCDHA8, THOC7, and TYW5) reached transcriptome-wide significance (TWS) level. These findings were confirmed by an independent integrative approach (i.e. Sherlock). We further conducted conditional analyses and identified the potential risk genes that driven the TWAS association signal in each locus. Gene expression analysis showed that several TWS genes (including CORO7, DDAH2, DDHD2, ELAC2, GLT8D1, THOC7 and TYW5) were dysregulated in the dorsolateral prefrontal cortex of SCZ cases compared with controls. TWS genes were mainly expressed on the surface of glutamatergic neurons, GABAergic neurons, and microglia. Finally, SCZ cases had a substantially greater TWS genes-based polygenic risk (PRS) compared to controls, and we showed that fractional anisotropy of the cingulum-hippocampus mediates the influence of TWS genes PRS on SCZ. CONCLUSIONS: Our findings identified novel SCZ risk genes and highlighted the importance of the TWS genes in frontal-limbic dysfunctions in SCZ, indicating possible therapeutic targets.

5.
Brain ; 145(2): 770-786, 2022 04 18.
Article in English | MEDLINE | ID: mdl-34581804

ABSTRACT

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).


Subject(s)
Genome-Wide Association Study , Mixed Function Oxygenases/genetics , Schizophrenia , Animals , Asian People/genetics , Genetic Predisposition to Disease/genetics , Humans , Mice , Polymorphism, Single Nucleotide/genetics , Rats , Schizophrenia/genetics
6.
Anal Chem ; 94(50): 17587-17594, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36464815

ABSTRACT

Immunohistochemistry (IHC) using fluorescent probes provides high resolution with multiplexing capability, but the imaging contrast is limited by the brightness of the fluorescent probe and the intrinsic autofluorescence background from tissues. Herein, we improved the contrast by high-density labeling of long-lifetime lanthanide complexes and time-gated imaging. As the large (∼280 nm) Stokes shift of lanthanide complexes effectively prevents the issue of concentration quenching, we succeeded in conjugating seven europium complexes to an eight-arm hydrophilic poly(ethylene glycol) (PEG) linker for signal amplification with improved water solubility to the level of up to 10 mg/mL. Moreover, we demonstrated that both human epidermal growth factor receptor 2 (HER2) in a formalin-fixed paraffin-embedded (FFPE) tissue section and cytokeratin 18 (CK18) in a frozen section can be resolved with the enhanced contrast by 2-fold and 3-fold, respectively. Furthermore, we show that the PEGylation of multiple lanthanide complexes is compatible with tyramide signal amplification (TSA). This work suggests new opportunities for sensitive imaging of low-abundance biomarkers in a tissue matrix.


Subject(s)
Lanthanoid Series Elements , Humans , Lanthanoid Series Elements/chemistry , Immunohistochemistry , Europium/chemistry , Fluorescent Dyes , Polyethylene Glycols
7.
BMC Med ; 20(1): 68, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35168626

ABSTRACT

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.


Subject(s)
Genome-Wide Association Study , Parkinson Disease , Genetic Predisposition to Disease/genetics , Genomics , Humans , Parkinson Disease/genetics , Polymorphism, Single Nucleotide/genetics
8.
BMC Med ; 20(1): 169, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35527273

ABSTRACT

BACKGROUND: Identifying the causal genes at the risk loci and elucidating their roles in schizophrenia (SCZ) pathogenesis remain significant challenges. To explore risk variants associated with gene expression in the human brain and to identify genes whose expression change may contribute to the susceptibility of SCZ, here we report a comprehensive integrative study on SCZ. METHODS: We systematically integrated the genetic associations from a large-scale SCZ GWAS (N = 56,418) and brain expression quantitative trait loci (eQTL) data (N = 175) using a Bayesian statistical framework (Sherlock) and Summary data-based Mendelian Randomization (SMR). We also measured brain structure of 86 first-episode antipsychotic-naive schizophrenia patients and 152 healthy controls with the structural MRI. RESULTS: Both Sherlock (P = 3. 38 × 10-6) and SMR (P = 1. 90 × 10-8) analyses showed that TYW5 mRNA expression was significantly associated with risk of SCZ. Brain-based studies also identified a significant association between TYW5 protein abundance and SCZ. The single-nucleotide polymorphism rs203772 showed significant association with SCZ and the risk allele is associated with higher transcriptional level of TYW5 in the prefrontal cortex. We further found that TYW5 was significantly upregulated in the brain tissues of SCZ cases compared with controls. In addition, TYW5 expression was also significantly higher in neurons induced from pluripotent stem cells of schizophrenia cases compared with controls. Finally, combining analysis of genotyping and MRI data showed that rs203772 was significantly associated with gray matter volume of the right middle frontal gyrus and left precuneus. CONCLUSIONS: We confirmed that TYW5 is a risk gene for SCZ. Our results provide useful information toward a better understanding of the genetic mechanism of TYW5 in risk of SCZ.


Subject(s)
Mixed Function Oxygenases , Schizophrenia , Bayes Theorem , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Mixed Function Oxygenases/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Schizophrenia/diagnostic imaging , Schizophrenia/genetics
9.
Mov Disord ; 37(12): 2451-2456, 2022 12.
Article in English | MEDLINE | ID: mdl-36177513

ABSTRACT

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.


Subject(s)
Genome-Wide Association Study , Parkinson Disease , Humans , Quantitative Trait Loci/genetics , Mendelian Randomization Analysis , Parkinson Disease/genetics , Parkinson Disease/metabolism , Dopaminergic Neurons/metabolism , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide
10.
Mol Psychiatry ; 26(11): 6896-6911, 2021 11.
Article in English | MEDLINE | ID: mdl-33931730

ABSTRACT

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).


Subject(s)
Schizophrenia , Alleles , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Mutation, Missense/genetics , Polymorphism, Single Nucleotide/genetics , Schizophrenia/genetics , YY1 Transcription Factor/genetics , alpha-N-Acetylgalactosaminidase/genetics , alpha-N-Acetylgalactosaminidase/metabolism
11.
J Med Genet ; 58(10): 666-678, 2021 10.
Article in English | MEDLINE | ID: mdl-32900838

ABSTRACT

The association between NOTCH4 and schizophrenia has been repeatedly reported. However, the results from different genetic studies are inconsistent, and the role of NOTCH4 in schizophrenia pathogenesis remains unknown. Here, we provide convergent lines of evidence that support NOTCH4 as a schizophrenia risk gene. We first performed a meta-analysis and found that a genetic variant (rs2071287) in NOTCH4 was significantly associated with schizophrenia (a total of 125 848 subjects, p=8.31×10-17), with the same risk allele across all tested samples. Expression quantitative trait loci (eQTL) analysis showed that rs2071287 was significantly associated with NOTCH4 expression (p=1.08×10-14) in human brain tissues, suggesting that rs2071287 may confer schizophrenia risk through regulating NOTCH4 expression. Sherlock integrative analysis using a large-scale schizophrenia GWAS and eQTL data from human brain tissues further revealed that NOTCH4 was significantly associated with schizophrenia (p=4.03×10-7 in CMC dataset and p=3.06×10-6 in xQTL dataset), implying that genetic variants confer schizophrenia risk through modulating NOTCH4 expression. Consistently, we found that NOTCH4 was significantly downregulated in brains of schizophrenia patients compared with controls (p=2.53×10-3), further suggesting that dysregulation of NOTCH4 may have a role in schizophrenia. Finally, we showed that NOTCH4 regulates proliferation, self-renewal, differentiation and migration of neural stem cells, suggesting that NOTCH4 may confer schizophrenia risk through affecting neurodevelopment. Our study provides convergent lines of evidence that support the involvement of NOTCH4 in schizophrenia. In addition, our study also elucidates a possible mechanism for the role of NOTCH4 in schizophrenia pathogenesis.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Receptor, Notch4/genetics , Schizophrenia/epidemiology , Schizophrenia/genetics , Alleles , Animals , Brain/metabolism , Case-Control Studies , Cells, Cultured , Chromosome Mapping , Computational Biology/methods , Gene Expression , Gene Knockdown Techniques , Genetic Association Studies/methods , Genome-Wide Association Study , Humans , Mice , Mice, Knockout , Molecular Sequence Annotation , Neural Stem Cells/metabolism , Neurons/metabolism , Polymorphism, Single Nucleotide , Population Surveillance , Quantitative Trait Loci , Schizophrenia/diagnosis
12.
BMC Med ; 19(1): 177, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34380480

ABSTRACT

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.


Subject(s)
Genome-Wide Association Study , Schizophrenia , Case-Control Studies , Genetic Predisposition to Disease , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci , RNA-Binding Proteins , Schizophrenia/genetics , Transcriptome
13.
Mol Psychiatry ; 25(9): 1926-1945, 2020 09.
Article in English | MEDLINE | ID: mdl-32214206

ABSTRACT

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.


Subject(s)
Depressive Disorder, Major , Genome-Wide Association Study , Depressive Disorder, Major/genetics , Genetic Predisposition to Disease/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics
14.
Am J Med Genet B Neuropsychiatr Genet ; 186(1): 28-39, 2021 01.
Article in English | MEDLINE | ID: mdl-33522098

ABSTRACT

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).


Subject(s)
DNA-Binding Proteins/metabolism , Genome-Wide Association Study , Neural Stem Cells/pathology , Neuroblastoma/pathology , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Schizophrenia/pathology , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Humans , Macaca , Neural Stem Cells/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Phenotype , Schizophrenia/genetics , Schizophrenia/metabolism , Transcription Factors/genetics , Tumor Cells, Cultured
15.
Hum Genet ; 139(10): 1285-1297, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32385526

ABSTRACT

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.


Subject(s)
Databases, Genetic , Epigenesis, Genetic , Genetic Predisposition to Disease , Multifactorial Inheritance , Quantitative Trait Loci , Schizophrenia/genetics , DNA Copy Number Variations , DNA Methylation , Gene Ontology , Genome-Wide Association Study , Humans , Information Storage and Retrieval/methods , Polymorphism, Single Nucleotide , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Protein Interaction Mapping , Risk , Schizophrenia/diagnosis , Schizophrenia/physiopathology
16.
J Antimicrob Chemother ; 74(9): 2559-2565, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31203365

ABSTRACT

OBJECTIVES: To eliminate mcr-1-harbouring plasmids and MDR plasmids in clinical Escherichia coli isolates. METHODS: Plasmid pMBLcas9 expressing Cas9 was constructed and used to clone target single-guide RNAs (sgRNAs) for plasmid curing. The recombinant plasmid pMBLcas9-sgRNA was transferred by conjugation into two clinical E. coli isolates. The curing efficiency of different sgRNAs targeting conserved genes was tested. The elimination of targeted plasmids and the generation of transposase-mediated recombination of p14EC033a variants were characterized by PCR and DNA sequencing. RESULTS: In this study, four native plasmids in isolate 14EC033 and two native plasmids in isolate 14EC007 were successfully eliminated in a step-by-step manner using pMBLcas9. Moreover, two native plasmids in 14EC007 were simultaneously eliminated by tandemly cloning multiple sgRNAs in pMBLcas9, sensitizing 14EC007 to polymyxin and carbenicillin. In 14EC033 with two mcr-1-harbouring plasmids, IncI2 plasmid p14EC033a and IncX4 plasmid p14EC033b, a single mcr-1 sgRNA mediated the loss of p14EC033b and generated a mutant p14EC033a in which the mcr-1 gene was deleted. An insertion element, IS5, located upstream of mcr-1 in p14EC033a was responsible for transposase-mediated recombination, resulting in mcr-1 gene deletion instead of plasmid curing. CONCLUSIONS: CRISPR/Cas9 can be used to efficiently sensitize clinical isolates to antibiotics in vitro. For isolates with multiple plasmids, the CRISPR/Cas9 approach can either remove each plasmid in a stepwise manner or simultaneously remove multiple plasmids in one step. Moreover, this approach can be used to delete multiple gene copies by using only one sgRNA. However, caution must be exercised to avoid unwanted recombination events during genetic manipulation.


Subject(s)
Anti-Bacterial Agents/pharmacology , CRISPR-Cas Systems , Escherichia coli Infections/microbiology , Escherichia coli/genetics , Plasmids/genetics , Bacterial Proteins/genetics , Conjugation, Genetic , DNA Transposable Elements/genetics , Escherichia coli/drug effects , Humans , RNA, Guide, Kinetoplastida/genetics , Recombination, Genetic
18.
Biochem Genet ; 57(1): 117-128, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30073579

ABSTRACT

Chronic hepatitis B virus (HBV) infection is an important etiology for the development of hepatocellular carcinoma (HCC). Tumor necrosis factor-α-induced protein 3-interacting protein 1 (TNIP1) is linked to specific inflammatory diseases as a novel type of endogenous inflammatory regulator. However, presently, rare information is found about the association between TNIP1 polymorphisms and HBV-induced HCC risk. In this case control study, we genotyped four single nucleotide polymorphisms (SNPs) in TNIP1 gene in 248 HCC patients and 242 chronic HBV carriers using Sequenom Mass-ARRAY technology. Genetic model and haplotype analysis were performed to evaluate the association between candidate SNPs polymorphisms and HBV-induced HCC susceptibility using Pearson's χ2 test and unconditional logistic regression analysis. Overall, we found two risk alleles in TNIP1 for HBV-induced HCC in patients: the allele "G" of rs7708392 by genotype model ("G/C" vs. "C/C": OR 1.88, 95% CI 1.17-3, P = 0.009) and dominant model ("G/C-G/G" vs. "C/C": OR 1.69, 95% CI 1.08-2.65, P = 0.023), and the allele "C" of rs10036748 by genotype model ("C/T" vs. "T/T": OR 1.83, 95% CI 1.14-2.92, P = 0.012) and dominant model ("C/T-C/C" vs. "T/T": OR 1.65, 95% CI 1.05-2.59, P = 0.03). However, rs3792792 and rs4958881 polymorphisms didn't significantly correlate with the risk of HBV-induced HCC. Haplotype analysis showed no significant association between haplotypes and the HCC risk in HBV carriers. This study provides evidence for HBV-induced HCC susceptibility gene TNIP1 in the Chinese Han population.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease/genetics , Hepatitis B, Chronic/complications , Liver Neoplasms/genetics , Polymorphism, Single Nucleotide , Carcinoma, Hepatocellular/complications , Case-Control Studies , China/ethnology , Female , Humans , Linkage Disequilibrium , Liver Neoplasms/complications , Male , Middle Aged
20.
Hereditas ; 155: 16, 2018.
Article in English | MEDLINE | ID: mdl-29308060

ABSTRACT

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.


Subject(s)
Calcium Channels, T-Type/genetics , Genetic Predisposition to Disease , Schizophrenia/genetics , Alleles , Asian People/genetics , Calcium Signaling , Case-Control Studies , China , Genotype , Humans , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL