1
|
Lorincz-Comi N, Yang Y, Ajayakumar J, Mews M, Bermudez V, Bush W, Zhu X. HORNET: tools to find genes with causal evidence and their regulatory networks using eQTLs. BIOINFORMATICS ADVANCES 2025; 5:vbaf068. [PMID: 40270926 PMCID: PMC12014422 DOI: 10.1093/bioadv/vbaf068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/17/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Motivation Nearly two decades of genome-wide association studies (GWAS) have identify thousands of disease-associated genetic variants, but very few genes with evidence of causality. Recent methodological advances demonstrate that Mendelian randomization (MR) using expression quantitative loci (eQTLs) as instrumental variables can detect potential causal genes. However, existing MR approaches are not well suited to handle the complexity of eQTL GWAS data structure and so they are subject to bias, inflation, and incorrect inference. Results We present a whole-genome regulatory network analysis tool (HORNET), which is a comprehensive set of statistical and computational tools to perform genome-wide searches for causal genes using summary level GWAS data, i.e. robust to biases from multiple sources. Applying HORNET to schizophrenia, eQTL effects in the cerebellum were spread throughout the genome, and in the cortex were more localized to select loci. Availability and implementation Freely available at https://github.com/noahlorinczcomi/HORNET or Mac, Windows, and Linux users.
Collapse
Affiliation(s)
- Noah Lorincz-Comi
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Yihe Yang
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Jayakrishnan Ajayakumar
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Makaela Mews
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Valentina Bermudez
- Case Western Reserve University Department of Neurosciences, Cleveland, OH 44106, United States
| | - William Bush
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Xiaofeng Zhu
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| |
Collapse
|
2
|
Baidoe-Ansah D, Mirzapourdelavar H, Aleshin S, Schott BH, Seidenbecher C, Kaushik R, Dityatev A. Neurocan regulates axon initial segment organization and neuronal activity. Matrix Biol 2025; 136:22-35. [PMID: 39788215 DOI: 10.1016/j.matbio.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The neural extracellular matrix (ECM) accumulates in the form of perineuronal nets (PNNs), particularly around fast-spiking GABAergic interneurons in the cortex and hippocampus, but also around synapses and in association with the axon initial segments (AIS) and nodes of Ranvier. Increasing evidence highlights the role of Neurocan (Ncan), a brain-specific component of ECM, in the pathophysiology of neuropsychiatric disorders like bipolar disorder and schizophrenia. Ncan localizes at PNNs, perisynaptically, and at the nodes of Ranvier and the AIS, highlighting its potential role in regulating axonal excitability. Here, we used knockdown and knockout approaches in mouse primary cortical neurons in combination with immunocytochemistry, Western blotting and electrophysiological techniques to characterize the role of Ncan in the organization of PNNs and AISs and regulation of neuronal activity. We found that reduced Ncan levels led to remodeling of PNNs around neurons via upregulation of aggrecan mRNA and protein levels, increased expression of activity-dependent c-Fos and FosB genes and elevated spontaneous synaptic activity. The latter correlated with increased levels of ankyrin-G in the AIS, particularly in excitatory neurons, and with the elevated expression of Nav1.6 channels. Our results suggest that Ncan regulates the expression of key proteins in PNNs and AISs and provide new insights into its role in fine-tuning neuronal functions.
Collapse
Affiliation(s)
- David Baidoe-Ansah
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Stepan Aleshin
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Björn Hendrik Schott
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Constanze Seidenbecher
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Rahul Kaushik
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
3
|
Jenkins PM, Bender KJ. Axon initial segment structure and function in health and disease. Physiol Rev 2025; 105:765-801. [PMID: 39480263 DOI: 10.1152/physrev.00030.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
At the simplest level, neurons are structured to integrate synaptic input and perform computational transforms on that input, converting it into an action potential (AP) code. This process, converting synaptic input into AP output, typically occurs in a specialized region of the axon termed the axon initial segment (AIS). The AIS, as its name implies, is often contained to the first section of axon abutted to the soma and is home to a dizzying array of ion channels, attendant scaffolding proteins, intracellular organelles, extracellular proteins, and, in some cases, synapses. The AIS serves multiple roles as the final arbiter for determining if inputs are sufficient to evoke APs, as a gatekeeper that physically separates the somatodendritic domain from the axon proper, and as a regulator of overall neuronal excitability, dynamically tuning its size to best suit the needs of parent neurons. These complex roles have received considerable attention from experimentalists and theoreticians alike. Here, we review recent advances in our understanding of the AIS and its role in neuronal integration and polarity in health and disease.
Collapse
Affiliation(s)
- Paul M Jenkins
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Kevin J Bender
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, California, United States
| |
Collapse
|
4
|
Cai N, Verhulst B, Andreassen OA, Buitelaar J, Edenberg HJ, Hettema JM, Gandal M, Grotzinger A, Jonas K, Lee P, Mallard TT, Mattheisen M, Neale MC, Nurnberger JI, Peyrot WJ, Tucker-Drob EM, Smoller JW, Kendler KS. Assessment and ascertainment in psychiatric molecular genetics: challenges and opportunities for cross-disorder research. Mol Psychiatry 2025; 30:1627-1638. [PMID: 39730880 PMCID: PMC11919726 DOI: 10.1038/s41380-024-02878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/07/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
Psychiatric disorders are highly comorbid, heritable, and genetically correlated [1-4]. The primary objective of cross-disorder psychiatric genetics research is to identify and characterize both the shared genetic factors that contribute to convergent disease etiologies and the unique genetic factors that distinguish between disorders [4, 5]. This information can illuminate the biological mechanisms underlying comorbid presentations of psychopathology, improve nosology and prediction of illness risk and trajectories, and aid the development of more effective and targeted interventions. In this review we discuss how estimates of comorbidity and identification of shared genetic loci between disorders can be influenced by how disorders are measured (phenotypic assessment) and the inclusion or exclusion criteria in individual genetic studies (sample ascertainment). Specifically, the depth of measurement, source of diagnosis, and time frame of disease trajectory have major implications for the clinical validity of the assessed phenotypes. Further, biases introduced in the ascertainment of both cases and controls can inflate or reduce estimates of genetic correlations. The impact of these design choices may have important implications for large meta-analyses of cohorts from diverse populations that use different forms of assessment and inclusion criteria, and subsequent cross-disorder analyses thereof. We review how assessment and ascertainment affect genetic findings in both univariate and multivariate analyses and conclude with recommendations for addressing them in future research.
Collapse
Affiliation(s)
- Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, Neuherberg, Germany
- Computational Health Centre, Helmholtz Munich, Neuherberg, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Brad Verhulst
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, USA
| | - Ole A Andreassen
- Centre of Precision Psychiatry, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Karakter Child and Adolescent University Center, Nijmegen, The Netherlands
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John M Hettema
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael Gandal
- Departments of Psychiatry and Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Grotzinger
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Katherine Jonas
- Department of Psychiatry & Behavioral Health, Stony Brook University, Stony Brook, NY, USA
| | - Phil Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Travis T Mallard
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Manuel Mattheisen
- Department of Community Health and Epidemiology and Faculty of Computer Science, Dalhousie University, Halifax, NS, Canada
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital of Munich, Munich, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - John I Nurnberger
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wouter J Peyrot
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Public Health, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Jordan W Smoller
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenneth S Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
5
|
Byrne RAJ, Nimmo J, Torvell M, Carpanini SM, Daskoulidou N, Hughes TR, Noble LV, Veteleanu A, Watkins LM, Zelek WM, O'Donovan MC, Morgan BP. The schizophrenia-associated gene CSMD1 encodes a complement classical pathway inhibitor predominantly expressed by astrocytes and at synapses in mice and humans. Brain Behav Immun 2025; 127:287-302. [PMID: 40112933 DOI: 10.1016/j.bbi.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
CUB and sushi multiple domains 1 (CSMD1) is predominantly expressed in brain and robustly associated with schizophrenia risk; however, understanding of which cells express CSMD1 in brain and how it impacts risk is lacking. CSMD1 encodes a large transmembrane protein including fifteen tandem short consensus repeats (SCRs), resembling complement C3 convertase regulators. CSMD1 complement regulatory activity has been reported and mapped to SCR17-21. We expressed two SCR domains of CSMD1, SCR17-21 and SCR23-26, and characterised their complement regulatory activity using a panel of functional assays testing convertase and terminal pathway inhibition. Both domains inhibited the classical pathway C3 convertase by acting as factor I cofactors; neither domain caused any inhibition in alternative or terminal pathway assays. Novel anti-CSMD1 monoclonal antibodies cross-reactive with human and mouse CSMD1 were generated that detected endogenous CSMD1 in human and rodent brain; immunostaining showed predominantly astrocyte and synaptic localisation of CSMD1, the latter confirmed using isolated synapses. Using iPSC-derived cells, astrocyte expression was confirmed and expression on cortical neurons demonstrated. We show that CSMD1 is a classical pathway-specific complement regulator expressed predominantly on astrocytes, neurons, and synapses in human and mouse brain. These findings will help reveal the mechanism by which CSMD1 impacts schizophrenia risk.
Collapse
Affiliation(s)
- Robert A J Byrne
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK.
| | - Jacqui Nimmo
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Megan Torvell
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Sarah M Carpanini
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Lucy V Noble
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Aurora Veteleanu
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Lewis M Watkins
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK
| | - Michael C O'Donovan
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK
| | - Bryan Paul Morgan
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; UK Dementia Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales CF24 4HQ, UK; Division of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, Wales CF14 4XN, UK.
| |
Collapse
|
6
|
Ji L, Huang B, Ren D, Wei X, Liu L, Bi Y, Li Z, Yuan F, Han K, Li K, Yang F, Li X, Yu T, Shi Y, He L, Lu Q, He G. Identification of Schizophrenia-Risk Regulatory Variant rs1399178 in the Non-coding Region With Its Impact on NRF1 Binding. CNS Neurosci Ther 2025; 31:e70275. [PMID: 40019038 PMCID: PMC11868986 DOI: 10.1111/cns.70275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/28/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
AIMS The challenges in identifying functional variants from genome-wide association studies (GWAS) and unraveling regulatory mechanisms in schizophrenia research persist, particularly in intronic regions. A non-coding regulatory variant, rs1399178, associated with schizophrenia risk, is identified and its impact on NRF1 binding is investigated. METHODS This study focuses on schizophrenia GWAS risk loci, using functional genomics, expression analyses and structural analysis to identify 736 schizophrenia risk single-nucleotide polymorphisms (SNPs) that disrupt transcription factor (TF) binding. RESULTS Among these SNPs, rs1399178 stands out as a bifunctional intergenic SNP that can switch between acting as a promoter and an enhancer, potentially influencing MLH1 and LRRFIP2 expression via expression quantitative trait loci analysis. Importantly, mutation of the G allele of rs1399178 to A significantly diminishes its binding affinity to nuclear respiratory factor 1 (NRF1). Structural analysis provides further insight into this alteration in the protein-nucleic acid complex formation. CONCLUSION Based on our data, a model is proposed in which rs1399178 confers schizophrenia risk by modifying NRF1 binding profiles, thereby regulating the abundance of target genes through promoter-enhancer switching. This study provides novel insights into the regulatory mechanisms of schizophrenia risk variants, highlighting the intricate nature of genetic interactions and potential therapeutic targets.
Collapse
Affiliation(s)
- Lei Ji
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bojian Huang
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Decheng Ren
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaoxi Wei
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liangjie Liu
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Bi
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhiqiang Li
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio‐X Institutes)Qingdao UniversityQingdaoChina
| | - Fan Yuan
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ke Han
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Keyi Li
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fengping Yang
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xingwang Li
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tao Yu
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi Shi
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lin He
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qing Lu
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- Department of Otorhinolaryngology‐Head and Neck SurgeryChongqing General HospitalChongqingChina
- Ear InstituteShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Guang He
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
7
|
Dang X, Teng Z, Yang Y, Li W, Liu J, Hui L, Zhou D, Gong D, Dai SS, Li Y, Li X, Lv L, Zeng Y, Yuan Y, Ma X, Liu Z, Li T, Luo XJ. Gene-level analysis reveals the genetic aetiology and therapeutic targets of schizophrenia. Nat Hum Behav 2025; 9:609-624. [PMID: 39753749 DOI: 10.1038/s41562-024-02091-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/18/2024] [Indexed: 03/27/2025]
Abstract
Genome-wide association studies (GWASs) have reported multiple risk loci for schizophrenia (SCZ). However, the majority of the associations were from populations of European ancestry. Here we conducted a large-scale GWAS in Eastern Asian populations (29,519 cases and 44,392 controls) and identified ten Eastern Asian-specific risk loci, two of which have not been previously reported. A further cross-ancestry GWAS meta-analysis (96,806 cases and 492,818 controls) including populations from diverse ancestries identified 61 previously unreported risk loci. Systematic variant-level analysis, including fine mapping, functional genomics and expression quantitative trait loci, prioritized potential causal variants. Gene-level analyses, including transcriptome-wide association study, proteome-wide association study and Mendelian randomization, nominated the potential causal genes. By integrating evidence from layers of different analyses, we prioritized the most plausible causal genes for SCZ, such as ACE, CNNM2, SNAP91, ABCB9 and GATAD2A. Finally, drug repurposing showed that ACE, CA14, MAPK3 and MAPT are potential therapeutic targets for SCZ. Our study not only showed the power of cross-ancestry GWAS in deciphering the genetic aetiology of SCZ, but also uncovered new genetic risk loci, potential causal variants and genes and therapeutic targets for SCZ.
Collapse
Affiliation(s)
- Xinglun Dang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Zhaowei Teng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Kunming, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Jiewei Liu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, China
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou, China
| | - Dongsheng Zhou
- Department of Psychiatry, Affiliated Kangning Hospital of Ningbo University (Ningbo Kangning Hospital), Ningbo, China
| | - Daohua Gong
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Shan-Shan Dai
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yifan Li
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Xingxing Li
- Department of Psychiatry, Affiliated Kangning Hospital of Ningbo University (Ningbo Kangning Hospital), Ningbo, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Yong Zeng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Kunming, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Xiancang Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Tao Li
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xiong-Jian Luo
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China.
| |
Collapse
|
8
|
Bhattacharyya U, John J, Lencz T, Lam M. Dissecting Schizophrenia Biology Using Pleiotropy With Cognitive Genomics. Biol Psychiatry 2025:S0006-3223(25)00989-8. [PMID: 39993652 DOI: 10.1016/j.biopsych.2025.02.890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/20/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Given the increasingly large number of loci discovered by psychiatric genome-wide association studies (GWASs), specification of the key biological pathways that underlie these loci has become a priority for the field. We have previously leveraged the pleiotropic genetic relationships between schizophrenia (SCZ) and 2 cognitive phenotypes (educational attainment and cognitive task performance) to differentiate 2 subsets of illness-relevant single nucleotide polymorphisms (SNPs): 1) those with concordant alleles, which are associated with reduced cognitive performance and educational attainment and increased SCZ risk, and 2) those with discordant alleles, which are linked to reduced educational and/or cognitive levels but lower SCZ susceptibility. METHODS In the current study, we extended our prior work, utilizing larger input GWAS datasets and a more powerful statistical approach to pleiotropic meta-analysis, the pleiotropic locus exploration and interpretation using optimal test (PLEIO). RESULTS Our pleiotropic meta-analysis of SCZ and the 2 cognitive phenotypes revealed 768 significant pleiotropic loci (166 novel). Among these, 347 loci harbored concordant SNPs, 270 encompassed discordant SNPs, and 151 dual loci contained concordant and discordant SNPs. Competitive gene-set analysis using MAGMA linked concordant SNP loci with neurodevelopmental pathways (e.g., neurogenesis), whereas discordant loci were associated with mature neuronal synaptic functions. These distinctions were also observed in BrainSpan analysis of temporal enrichment patterns across developmental periods, with concordant loci containing more prenatally expressed genes than discordant loci. Dual loci were enriched for genes related to messenger RNA translation initiation, which represents a novel finding in the SCZ literature. CONCLUSIONS Pleiotropic analysis permits not only enhanced statistical power for locus discovery but also the ability to parse distinct biological processes associated with endophenotypes.
Collapse
Affiliation(s)
- Upasana Bhattacharyya
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, New York; Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, New York
| | - Jibin John
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, New York; Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, New York
| | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, New York; Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, New York; Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York.
| | - Max Lam
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, New York; Institute of Mental Health, Singapore; Department of Population and Global Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| |
Collapse
|
9
|
Khan R, Turner A, Berk M, Walder K, Rossell S, Guerin AA, Kim JH. Genes, Cognition, and Their Interplay in Methamphetamine Use Disorder. Biomolecules 2025; 15:306. [PMID: 40001609 PMCID: PMC11852989 DOI: 10.3390/biom15020306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Methamphetamine use disorder is a pressing global health issue, often accompanied by significant cognitive deficits that impair daily functioning and quality of life and complicate treatment. Emerging evidence highlights the potential role of genetic factors in methamphetamine use disorder, particularly in association with cognitive function. This review examines the key genetic and cognitive dimensions and their interplay in methamphetamine use disorder. There is converging evidence from several studies that genetic polymorphisms in BDNF, FAAH, SLC18A1, and SLC18A2 are associated with protection against or susceptibility to the disorder. In addition, people with methamphetamine use disorder consistently displayed impairments in cognitive flexibility and inhibitory control compared with people without the disorder. These cognitive domains were associated with reactivity to methamphetamine cues that were positively correlated with total years of methamphetamine use history. Emerging research also suggests that inhibitory control is negatively correlated with lower blood FAAH mRNA levels, while cognitive flexibility positively correlates with higher blood SLC18A2 mRNA levels, highlighting how genetic and cognitive dimensions interact in methamphetamine use disorder. We also include some future directions, emphasizing potential personalized therapeutic strategies that integrate genetic and cognitive insights. By drawing attention to the interplay between genes and cognition, we hope to advance our understanding of methamphetamine use disorder and inform the development of targeted interventions.
Collapse
Affiliation(s)
- Ramisha Khan
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (R.K.); (A.T.); (M.B.); (K.W.)
| | - Alyna Turner
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (R.K.); (A.T.); (M.B.); (K.W.)
| | - Michael Berk
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (R.K.); (A.T.); (M.B.); (K.W.)
| | - Ken Walder
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (R.K.); (A.T.); (M.B.); (K.W.)
| | - Susan Rossell
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC 3122, Australia;
| | - Alexandre A. Guerin
- Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia;
- Orygen, Melbourne, VIC 3052, Australia
| | - Jee Hyun Kim
- IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (R.K.); (A.T.); (M.B.); (K.W.)
| |
Collapse
|
10
|
Li R, Taliun SAG, Liao K, Flickinger M, Sobell JL, Genovese G, Locke AE, Chiu RR, LeFaive J, Wang J, Martins T, Chapman S, Neumann A, Handsaker RE, Arnett DK, Barnes KC, Boerwinkle E, Braff D, Cade BE, Fornage M, Gibbs RA, Hoth KF, Hou L, Kooperberg C, Loos RJ, Metcalf GA, Montgomery CG, Morrison AC, Qin ZS, Redline S, Reiner AP, Rich SS, Rotter JI, Taylor KD, Viaud-Martinez KA, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, Genomic Psychiatry Cohort investigators, Bigdeli TB, Gabriel S, Zollner S, Smith AV, Abecasis G, McCarroll S, Pato MT, Pato CN, Boehnke M, Knowles J, Kang HM, Ophoff RA, Ernst J, Scott LJ. Whole genome sequence-based association analysis of African American individuals with bipolar disorder and schizophrenia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.27.24319111. [PMID: 39763555 PMCID: PMC11703280 DOI: 10.1101/2024.12.27.24319111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
In studies of individuals of primarily European genetic ancestry, common and low-frequency variants and rare coding variants have been found to be associated with the risk of bipolar disorder (BD) and schizophrenia (SZ). However, less is known for individuals of other genetic ancestries or the role of rare non-coding variants in BD and SZ risk. We performed whole genome sequencing of African American individuals: 1,598 with BD, 3,295 with SZ, and 2,651 unaffected controls (InPSYght study). We increased power by incorporating 14,812 jointly called psychiatrically unscreened ancestry-matched controls from the Trans-Omics for Precision Medicine (TOPMed) Program for a total of 17,463 controls. To identify variants and sets of variants associated with BD and/or SZ, we performed single-variant tests, gene-based tests for singleton protein truncating variants, and rare and low-frequency variant annotation-based tests with conservation and universal chromatin states and sliding windows. We found suggestive evidence of BD association with single-variants on chromosome 18 and of lower BD risk associated with rare and low-frequency variants on chromosome 11 in a region with multiple BD GWAS loci, using a sliding window approach. We also found that chromatin and conservation state tests can be used to detect differential calling of variants in controls sequenced at different centers and to assess the effectiveness of sequencing metric covariate adjustments. Our findings reinforce the need for continued whole genome sequencing in additional samples of African American individuals and more comprehensive functional annotation of non-coding variants.
Collapse
Affiliation(s)
- Runjia Li
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA
- These authors contributed equally: Runjia Li, Sarah A. Gagliano Taliun
| | - Sarah A. Gagliano Taliun
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute Research Centre, Montreal, Quebec, Canada
- These authors contributed equally: Runjia Li, Sarah A. Gagliano Taliun
- Senior authors
| | - Kevin Liao
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Genomics plc, Oxford, UK
| | - Matthew Flickinger
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Janet L. Sobell
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam E. Locke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Rebeca Rothwell Chiu
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Jonathon LeFaive
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Jiongming Wang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Taylor Martins
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Arizona Department of Health Services, Phoenix, AZ, USA
| | - Sinéad Chapman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anna Neumann
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert E. Handsaker
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Gentics, Harvard Medical School, Boston, MA, USA
| | - Donna K. Arnett
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Kathleen C. Barnes
- Departments of Medicine & Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric Boerwinkle
- Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - David Braff
- Department of Psychiatry, University of California, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Brian E. Cade
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Richard A. Gibbs
- Baylor College of Medicine Human Genome Sequencing Center, Department of Molecular and Human Genetics, Houston, TX, USA
| | - Karin F. Hoth
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ruth J.F. Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ginger A. Metcalf
- Baylor College of Medicine Human Genome Sequencing Center, Department of Molecular and Human Genetics, Houston, TX, USA
| | | | - Alanna C. Morrison
- Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Zhaohui S. Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Stephen S. Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | | | | | - Tim B. Bigdeli
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- VA New York Harbor Healthcare System, Brooklyn, NY, USA
| | - Stacey Gabriel
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sebastian Zollner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Albert V. Smith
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Goncalo Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Steve McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 260, Boston, MA, USA
| | - Michele T. Pato
- Department of Psychiatry, Rutgers University, New Brunswick, NJ, USA
| | - Carlos N. Pato
- Department of Psychiatry, Rutgers University, New Brunswick, NJ, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - James Knowles
- Human Genetics Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Hyun Min Kang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Roel A. Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Senior authors
| | - Jason Ernst
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA
- Department of Computational Medicine, University of California, Los Angeles, CA, USA
- Computer Science Department, University of California, Los Angeles, CA, USA
- Senior authors
| | - Laura J. Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Senior authors
| |
Collapse
|
11
|
Liang L, Zhang S, Wang Z, Zhang H, Li C, Duhe AC, Sun X, Zhong X, Kozlova A, Jamison B, Wood W, Pang ZP, Sanders AR, He X, Duan J. Single-cell multiomics of neuronal activation reveals context-dependent genetic control of brain disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638682. [PMID: 40027724 PMCID: PMC11870544 DOI: 10.1101/2025.02.17.638682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Despite hundreds of genetic risk loci identified for neuropsychiatric disorders (NPD), most causal variants/genes remain unknown. A major hurdle is that disease risk variants may act in specific biological contexts, e.g., during neuronal activation, which is difficult to study in vivo at the population level. Here, we conducted a single-cell multiomics study of neuronal activation (stimulation) in human iPSC-induced excitatory and inhibitory neurons from 100 donors, and uncovered abundant neuronal stimulation-specific causal variants/genes for NPD. We surveyed NPD-relevant transcriptomic and epigenomic landscape of neuronal activation and identified thousands of genetic variants associated with activity-dependent gene expression (i.e., eQTL) and chromatin accessibility (i.e., caQTL). These caQTL explained considerably larger proportions of NPD heritability than the eQTL. Integrating the multiomic data with GWAS further revealed NPD risk variants/genes whose effects were only detected upon stimulation. Interestingly, multiple lines of evidence support a role of activity-dependent cholesterol metabolism in NPD. Our work highlights the power of cell stimulation to reveal context-dependent "hidden" genetic effects.
Collapse
Affiliation(s)
- Lifan Liang
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, USA
| | - Zicheng Wang
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
| | - Chuxuan Li
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra C. Duhe
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
| | - Xiaotong Sun
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaoyuan Zhong
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Alena Kozlova
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
| | - Brendan Jamison
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
| | - Whitney Wood
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
| | - Zhiping P. Pang
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Alan R. Sanders
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, USA
| | - Xin He
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
12
|
Duncan LE, Li T, Salem M, Li W, Mortazavi L, Senturk H, Shahverdizadeh N, Vesuna S, Shen H, Yoon J, Wang G, Ballon J, Tan L, Pruett BS, Knutson B, Deisseroth K, Giardino WJ. Mapping the cellular etiology of schizophrenia and complex brain phenotypes. Nat Neurosci 2025; 28:248-258. [PMID: 39833308 PMCID: PMC11802450 DOI: 10.1038/s41593-024-01834-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/29/2024] [Indexed: 01/22/2025]
Abstract
Psychiatric disorders are multifactorial and effective treatments are lacking. Probable contributing factors to the challenges in therapeutic development include the complexity of the human brain and the high polygenicity of psychiatric disorders. Combining well-powered genome-wide and brain-wide genetics and transcriptomics analyses can deepen our understanding of the etiology of psychiatric disorders. Here, we leverage two landmark resources to infer the cell types involved in the etiology of schizophrenia, other psychiatric disorders and informative comparison of brain phenotypes. We found both cortical and subcortical neuronal associations for schizophrenia, bipolar disorder and depression. These cell types included somatostatin interneurons, excitatory neurons from the retrosplenial cortex and eccentric medium spiny-like neurons from the amygdala. In contrast we found T cell and B cell associations with multiple sclerosis and microglial associations with Alzheimer's disease. We provide a framework for a cell-type-based classification system that can lead to drug repurposing or development opportunities and personalized treatments. This work formalizes a data-driven, cellular and molecular model of complex brain disorders.
Collapse
Affiliation(s)
- Laramie E Duncan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| | - Tayden Li
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Madeleine Salem
- Vice Provost for Undergraduate Education, Stanford University, Stanford, CA, USA
| | - Will Li
- Vice Provost for Undergraduate Education, Stanford University, Stanford, CA, USA
| | - Leili Mortazavi
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Hazal Senturk
- Department of Computer Science, University of San Francisco, San Francisco, CA, USA
| | - Naghmeh Shahverdizadeh
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sam Vesuna
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Hanyang Shen
- Department of Epidemiology, Stanford University, Stanford, CA, USA
| | - Jong Yoon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Gordon Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jacob Ballon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Longzhi Tan
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | | | - Brian Knutson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - William J Giardino
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
13
|
Martinez-Sanchez M, Skarnes W, Jain A, Vemula S, Sun L, Rockowitz S, Whitman MC. Chromosome 4 Duplication Associated with Strabismus Leads to Gene Expression Changes in iPSC-Derived Cortical Neurons. Genes (Basel) 2025; 16:80. [PMID: 39858627 PMCID: PMC11764630 DOI: 10.3390/genes16010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Strabismus is the most common ocular disorder of childhood. Three rare, recurrent genetic duplications have been associated with both esotropia and exotropia, but the mechanisms by which they contribute to strabismus are unknown. This work aims to investigate the mechanisms of the smallest of the three, a 23 kb duplication on chromosome 4 (hg38|4:25,554,985-25,578,843). METHODS Using CRISPR and bridging oligos, we introduced the duplication into the Kolf2.1J iPSC line. We differentiated the parent line and the line with the duplication into cortical neurons using a three-dimensional differentiation protocol, and performed bulk RNASeq on neural progenitors (day 14) and differentiated neurons (day 63). RESULTS We successfully introduced the duplication into Kolf2.1J iPSCs by nucleofecting a bridging oligo for the newly formed junction along with cas9 ribonucleoparticles. We confirmed that the cells had a tandem duplication without inversion or deletion. The parent line and the line with the duplication both differentiated into neurons reliably. There were a total of 37 differentially expressed genes (DEGs) at day 63, 25 downregulated and 12 upregulated. There were 55 DEGs at day 14, 18 of which were also DEGs at day 63. The DEGs included a number of protocadherins, several genes involved in neuronal development, including SLITRK2, CSMD1, and VGF, and several genes of unknown function. CONCLUSIONS A copy number variant (CNV) that confers risk for strabismus affects gene expression of several genes involved in neural development, highlighting that strabismus most likely results from abnormal neural development, and identifying several new genes and pathways for further research into the pathophysiology of strabismus.
Collapse
Affiliation(s)
- Mayra Martinez-Sanchez
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA 02115, USA; (M.M.-S.); (S.V.)
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - William Skarnes
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA;
| | - Ashish Jain
- Research Computing, Department of Information Technology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.J.); (L.S.); (S.R.)
| | - Sampath Vemula
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA 02115, USA; (M.M.-S.); (S.V.)
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Liang Sun
- Research Computing, Department of Information Technology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.J.); (L.S.); (S.R.)
| | - Shira Rockowitz
- Research Computing, Department of Information Technology, Boston Children’s Hospital, Boston, MA 02115, USA; (A.J.); (L.S.); (S.R.)
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Mary C. Whitman
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA 02115, USA; (M.M.-S.); (S.V.)
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
14
|
Sager REH, North HF, Weissleder C, Clearwater MS, Walker AK, Fullerton JM, Webster MJ, Shannon Weickert C. Divergent changes in complement pathway gene expression in schizophrenia and bipolar disorder: Links to inflammation and neurogenesis in the subependymal zone. Schizophr Res 2025; 275:25-34. [PMID: 39616737 DOI: 10.1016/j.schres.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/20/2024] [Accepted: 11/21/2024] [Indexed: 01/23/2025]
Abstract
Deficits in neurogenesis markers in the subependymal zone (SEZ) are associated with elevated inflammation in schizophrenia and bipolar disorder. However, the extent to which complement factors are also changed in the SEZ of these major psychiatric disorders and their impact on neurogenesis remains poorly understood. We extracted RNA from the SEZ of 93 brains, including controls (n = 32), schizophrenia (n = 32), and bipolar disorder (n = 29) cases. Quantitative RT-PCR measured 13 complement transcripts encoding initiators, convertases, effectors or inhibitors. Differences in abundance were analysed by diagnosis and inflammatory subgroups (high- or low-inflammation), which were previously defined by SEZ cytokine and inflammation marker expression. Complement mRNAs C1QA (p = 0.011), C1QB (p < 0.001), C1R (p = 0.027), and Factor B (p = 0.025) were increased in high-inflammation schizophrenia versus low-inflammation controls. Conversely, high-inflammation bipolar cases had decreased C1QC (p = 0.011) and C3 (p = 0.003). Complement mRNAs C1R (SCZ, p = 0.010; BD, p = 0.047), C1S (SCZ, p = 0.026; BD, p = 0.017), and Factor B (BD, p = 0.025) were decreased in low-inflammation schizophrenia and bipolar subgroups versus low-inflammation controls. Complement inhibitors varied by subgroup: Factor H was increased in high-inflammation schizophrenia (p < 0.001), and CD59 in high-inflammation bipolar disorder (p = 0.020). Complement activator and inhibitor mRNAs were positively correlated with quiescent neural stem cell marker GFAPD (q < 0.05) but negatively with immature neuron markers DLX6-AS1 (q < 0.05) and DCX (q < 0.05). These findings suggest altered complement cascade expression in the SEZ in high- and low-inflammation schizophrenia and bipolar disorder, with opposite directional changes suggesting distinct molecular pathology. Complement activation may promote stem cell quiescence and reduce differentiation or survival of newborn neurons.
Collapse
Affiliation(s)
- Rachel E H Sager
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Hayley F North
- Neuroscience Research Australia, Randwick, NSW, Australia; Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Christin Weissleder
- Mechanism and therapy for genetic brain diseases, Institut Imagine, Paris, France
| | - Misaki S Clearwater
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Adam K Walker
- Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, NSW, Australia
| | - Janice M Fullerton
- Neuroscience Research Australia, Randwick, NSW, Australia; School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Maree J Webster
- Stanley Medical Research Institute, 9800 Medical Center Drive, Rockville, MD, USA
| | - Cynthia Shannon Weickert
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA; Neuroscience Research Australia, Randwick, NSW, Australia; Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
15
|
Ding X, Wu Y, Vainshtein A, Rodriguez V, Ricco E, Okoh JT, Liu Y, Kraushaar DC, Peles E, Rasband MN. Age-dependent regulation of axoglial interactions and behavior by oligodendrocyte AnkyrinG. Nat Commun 2024; 15:10865. [PMID: 39738113 PMCID: PMC11686269 DOI: 10.1038/s41467-024-55209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
The bipolar disorder (BD) risk gene ANK3 encodes the scaffolding protein AnkyrinG (AnkG). In neurons, AnkG regulates polarity and ion channel clustering at axon initial segments and nodes of Ranvier. Disruption of neuronal AnkG causes BD-like phenotypes in mice. During development, AnkG is also expressed at comparable levels in oligodendrocytes and facilitates the efficient assembly of paranodal junctions. However, the physiological roles of glial AnkG in the mature nervous system, and its contributions to BD-like phenotypes, remain unexplored. Here, we show that oligodendroglia-specific AnkG conditional knockout results in destabilization of axoglial interactions in aged but not young adult mice. In addition, these mice exhibit significant histological, electrophysiological, and behavioral pathophysiologies. Unbiased translatomic profiling reveals potential compensatory machineries. These results highlight the functions of glial AnkG in maintaining proper axoglial interactions throughout aging and suggest a contribution of glial AnkG to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna Vainshtein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Victoria Rodriguez
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Emily Ricco
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - James T Okoh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yanhong Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daniel C Kraushaar
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Lippincott MF, Schafer EC, Hindman AA, He W, Brauner R, Delaney A, Grinspon R, Hall JE, Hirschhorn JN, McElreavey K, Palmert MR, Rey R, Seminara SB, Salem RM, Chan YM. Contributions of Common Genetic Variants to Constitutional Delay of Puberty and Idiopathic Hypogonadotropic Hypogonadism. J Clin Endocrinol Metab 2024; 110:e61-e67. [PMID: 38477512 PMCID: PMC11651688 DOI: 10.1210/clinem/dgae166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
CONTEXT Constitutional delay of puberty (CDP) is highly heritable, but the genetic basis for CDP is largely unknown. Idiopathic hypogonadotropic hypogonadism (IHH) can be caused by rare genetic variants, but in about half of cases, no rare-variant cause is found. OBJECTIVE To determine whether common genetic variants that influence pubertal timing contribute to CDP and IHH. DESIGN Case-control study. PARTICIPANTS 80 individuals with CDP; 301 with normosmic IHH, and 348 with Kallmann syndrome (KS); control genotyping data from unrelated studies. MAIN OUTCOME MEASURES Polygenic scores (PGS) based on genome-wide association studies for timing of male pubertal hallmarks and age at menarche (AAM). RESULTS The CDP cohort had higher PGS for male pubertal hallmarks and for AAM compared to controls (for male hallmarks, Cohen's d = 0.67, P = 1 × 10-10; for AAM, d = 0.85, P = 1 × 10-16). The normosmic IHH cohort also had higher PGS for male hallmarks compared to controls, but the difference was smaller (male hallmarks d = 0.20, P = .003; AAM d = 0.10, P = .055). No differences were seen for the KS cohort compared to controls (male hallmarks d = 0.05, P = .45; AAM d = 0.03, P = .56). CONCLUSION Common genetic variants that influence pubertal timing in the general population contribute strongly to the genetics of CDP, weakly to normosmic IHH, and potentially not at all to KS. These findings demonstrate that the common-variant genetics of CDP and normosmic IHH are largely but not entirely distinct.
Collapse
Affiliation(s)
- Margaret F Lippincott
- Harvard Center for Reproductive Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Departments of Medicine (M.F.L., S.B.S.), Pediatrics (J.N.H., Y.-M.C.), and Genetics (J.N.H.), Harvard Medical School, Boston, MA 02115, USA
| | - Evan C Schafer
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anna A Hindman
- Harvard Center for Reproductive Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Wen He
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Raja Brauner
- Unité d'Endocrinologie Pédiatrique et Troubles de la Croissance, Hôpital Fondation Adolphe de Rothschild and Université Paris Cité, 75019 Paris, France
| | - Angela Delaney
- Division of Endocrinology, Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Romina Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET—FEI—División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Janet E Hall
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Joel N Hirschhorn
- Departments of Medicine (M.F.L., S.B.S.), Pediatrics (J.N.H., Y.-M.C.), and Genetics (J.N.H.), Harvard Medical School, Boston, MA 02115, USA
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Programs in Medical and Population Genetics (J.N.H., S.B.S., Y.-M.C.) and Metabolism (J.N.H.), Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kenneth McElreavey
- Human Developmental Genetics, CNRS UMR3738, Institut Pasteur, 75015 Paris, France
| | - Mark R Palmert
- Division of Endocrinology, Hospital for Sick Children, Departments of Pediatrics and Physiology, University of Toronto, Toronto, ON M5G 1E8, Canada
| | - Rodolfo Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET—FEI—División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Stephanie B Seminara
- Harvard Center for Reproductive Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Departments of Medicine (M.F.L., S.B.S.), Pediatrics (J.N.H., Y.-M.C.), and Genetics (J.N.H.), Harvard Medical School, Boston, MA 02115, USA
- Programs in Medical and Population Genetics (J.N.H., S.B.S., Y.-M.C.) and Metabolism (J.N.H.), Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rany M Salem
- Herbert Wertheim School of Public Health & Human Longevity Science, University of San Diego, La Jolla, CA 92093, USA
| | - Yee-Ming Chan
- Departments of Medicine (M.F.L., S.B.S.), Pediatrics (J.N.H., Y.-M.C.), and Genetics (J.N.H.), Harvard Medical School, Boston, MA 02115, USA
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Programs in Medical and Population Genetics (J.N.H., S.B.S., Y.-M.C.) and Metabolism (J.N.H.), Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
17
|
Millat MS, Roy J, Rahman MA, Aziz MA, Islam S, Chowdhury MMI, Barek MA, Hussain MS, Uddin MS, Siddiqui SA, Islam MS. Association of NOTCH4 and CYP2E1 Genetic Variants With Schizophrenia in the Bangladeshi Population: A Case-Control Study. Health Sci Rep 2024; 7:e70262. [PMID: 39698532 PMCID: PMC11652386 DOI: 10.1002/hsr2.70262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 10/03/2024] [Accepted: 11/23/2024] [Indexed: 12/20/2024] Open
Abstract
Background and Aims Schizophrenia (SCZ) is among the most persistent and devastating psychological problems. Different genetic polymorphisms are responsible for the predisposition of SCZ, and we screened NOTCH4 (rs2071287, rs204993) and CYP2E1 (rs2070673) polymorphisms in this study to find the connection with SCZ development. Methods We investigated a total of 420 samples (210 patients and 210 controls) and used the PCR-RFLP technique to genotype all SNPs. For statistical analyses, SPSS (version 25.0) was applied. Results In the case of NOTCH4 rs2071287, no evidence of a link was found in any genetic models, whereas NOTCH4 rs204993 and CYP2E1 rs2070673 showed a significant linkage in four genetic models with SCZ risk (for NOTCH4 rs204993, additive model 2: OR = 3.39, CI = 1.84-6.23, p = 0.0001; dominant: OR = 1.84, CI = 1.22-2.76, p = 0.0032; recessive: OR = 2.67, CI = 1.53-4.64, p = 0.0005; allelic: OR = 1.75, CI = 1.32-2.30, p = 0.0001 and for CYP2E1 rs2070673, additive model 2: OR = 0.39, CI = 0.22-0.69, p = 0.0013; recessive: OR = 0.45, CI = 0.29-0.68, p = 0.0002; overdominant: OR = 1.49, CI = 1.02-2.19, p = 0.0408; allelic: OR = 0.61, CI = 0.46-0.80, p = 0.0004). Conclusions The findings of our study suggest that the polymorphisms NOTCH4 rs204993 and CYP2E1 rs2070673 in the Bangladeshi ethnicity are connected to the risk of SCZ.
Collapse
Affiliation(s)
- Md. Shalahuddin Millat
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Joysree Roy
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Md. Atikur Rahman
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Md. Abdul Aziz
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Safiqul Islam
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | | | - Md Abdul Barek
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Md. Saddam Hussain
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Mohammad Sarowar Uddin
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Shafayet Ahmed Siddiqui
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| | - Mohammad Safiqul Islam
- Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
- Laboratory of Pharmacogenomics and Molecular Biology, Department of PharmacyNoakhali Science and Technology UniversitySonapurNoakhaliBangladesh
| |
Collapse
|
18
|
Kim B, Kim HA, Woo J, Lee HJ, Kim TK, Min H, Lee CJ, Im HI. Striatal Cholinergic Interneurons Control Physical Nicotine Withdrawal via Muscarinic Receptor Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402274. [PMID: 39491887 DOI: 10.1002/advs.202402274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/26/2024] [Indexed: 11/05/2024]
Abstract
Striatal cholinergic interneurons (ChIs) provide acetylcholine tone to the striatum and govern motor functions. Nicotine withdrawal elicits physical symptoms that dysregulate motor behavior. Here, the role of striatal ChIs in physical nicotine withdrawal is investigated. Mice under RNAi-dependent genetic inhibition of striatal ChIs (ChIGI) by suppressing the sodium channel subunit NaV1.1, lessening action potential generation and activity-dependent acetylcholine release is first generated. ChIGI markedly reduced the somatic signs of nicotine withdrawal without affecting other nicotine-dependent or striatum-associated behaviors. Multielectrode array (MEA) recording revealed that ChIGI reversed ex vivo nicotine-induced alterations in the number of neural population spikes in the dorsal striatum. Notably, the drug repurposing strategy revealed that a clinically-approved antimuscarinic drug, procyclidine, fully mimicked the therapeutic electrophysiological effects of ChIGI. Furthermore, both ChIGI and procyclidine prevented the nicotine withdrawal-induced reduction in striatal dopamine release in vivo. Lastly, therapeutic intervention with procyclidine dose-dependently diminished the physical signs of nicotine withdrawal. The data demonstrated that the striatal ChIs are a critical substrate of physical nicotine withdrawal and that muscarinic antagonism holds therapeutic potential against nicotine withdrawal.
Collapse
Affiliation(s)
- Baeksun Kim
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Han Ah Kim
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Junsung Woo
- Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul, 02792, Republic of Korea
| | - Hyeon-Jeong Lee
- Doping Control Center, KIST, Seoul, 02792, Republic of Korea
| | - Tae Kyoo Kim
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hophil Min
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- Doping Control Center, KIST, Seoul, 02792, Republic of Korea
| | - C Justin Lee
- Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul, 02792, Republic of Korea
| | - Heh-In Im
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| |
Collapse
|
19
|
Honda T, Kurita K, Arai Y, Pandey H, Sawa A, Furukubo-Tokunaga K. FMR1 genetically interacts with DISC1 to regulate glutamatergic synaptogenesis. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:112. [PMID: 39604386 PMCID: PMC11603133 DOI: 10.1038/s41537-024-00532-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024]
Abstract
Synaptic development and functions have been hypothesized as crucial mechanisms of diverse neuropsychiatric disorders. Studies in past years suggest that mutations in the fragile X mental retardation 1 (FMR1) are associated with diverse mental disorders including intellectual disability, autistic spectrum disorder, and schizophrenia. In this study, we have examined genetical interactions between a select set of risk factor genes using fruit flies to find that dfmr1, the Drosophila homolog of the human FMR1 gene, exhibits functional interactions with DISC1 in synaptic development. We show that DISC1 overexpression in the dfmr1null heterozygous background causes synaptic alterations at the larval neuromuscular junctions that are distinct from those in the wild-type background. Loss of dfmr1 modifies the DISC1 overexpression phenotype in synaptic formation, suppressing the formation of synapse boutons. Interaction between the two genes was further supported molecularly by the results that dfmr1 mutations suppress the DISC1-mediated upregulations of the postsynaptic expression of a glutamate receptor and the expression of ELKS/CAST protein, Bruchpilot, in presynaptic motoneurons. Moreover, DISC1 overexpression in the dfmr1null heterozygous background causes downregulation of a MAP1 family protein, Futsch. These results thus suggest an intriguing converging mechanism controlled by FMR1 and DISC1 in the developing glutamatergic synapses.
Collapse
Affiliation(s)
- Takato Honda
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusettes Institute of Technology (MIT), Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusettes General Hospital, Harvard Medical School, Boston, MA, USA.
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
| | - Kazuki Kurita
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yuko Arai
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Himani Pandey
- Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Akira Sawa
- Departments of Psychiatry, Neuroscience, Mental Health, Pharmacology, Biomedical Engineering and Genetic Medicine, Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Johns Hopkins Medicine, Baltimore, MD, USA
| | | |
Collapse
|
20
|
Arokiaraj CM, Leone MJ, Kleyman M, Chamessian A, Noh MC, Phan BN, Lopes BC, Corrigan KA, Cherupally VK, Yeramosu D, Franusich ME, Podder R, Lele S, Shiers S, Kang B, Kennedy MM, Chen V, Chen Z, Mathys H, Dum RP, Lewis DA, Qadri Y, Price TJ, Pfenning AR, Seal RP. Spatial, transcriptomic, and epigenomic analyses link dorsal horn neurons to chronic pain genetic predisposition. Cell Rep 2024; 43:114876. [PMID: 39453813 PMCID: PMC11801220 DOI: 10.1016/j.celrep.2024.114876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/07/2024] [Accepted: 09/30/2024] [Indexed: 10/27/2024] Open
Abstract
Key mechanisms underlying chronic pain occur within the dorsal horn. Genome-wide association studies (GWASs) have identified genetic variants predisposed to chronic pain. However, most of these variants lie within regulatory non-coding regions that have not been linked to spinal cord biology. Here, we take a multi-species approach to determine whether chronic pain variants impact the regulatory genomics of dorsal horn neurons. First, we generate a large rhesus macaque single-nucleus RNA sequencing (snRNA-seq) atlas and integrate it with available human and mouse datasets to produce a single unified, species-conserved atlas of neuron subtypes. Cellular-resolution spatial transcriptomics in mouse shows the precise laminar location of these neuron subtypes, consistent with our analysis of neuron-subtype-selective markers in macaque. Using this cross-species framework, we generate a mouse single-nucleus open chromatin atlas of regulatory elements that shows strong and selective relationships between the neuron-subtype-specific chromatin regions and variants from major chronic pain GWASs.
Collapse
Affiliation(s)
- Cynthia M Arokiaraj
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael J Leone
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael Kleyman
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Alexander Chamessian
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27708, USA; Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Myung-Chul Noh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - BaDoi N Phan
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Bettega C Lopes
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kelly A Corrigan
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Vijay Kiran Cherupally
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Deepika Yeramosu
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Michael E Franusich
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Riya Podder
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Sumitra Lele
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Byungsoo Kang
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Meaghan M Kennedy
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Viola Chen
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ziheng Chen
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Hansruedi Mathys
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Richard P Dum
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Yawar Qadri
- Department of Anesthesiology, Emory University, Atlanta, GA 30038, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Andreas R Pfenning
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| | - Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
21
|
Shi R, Chang X, Banaschewski T, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Martinot17, MLP, Artiges E, Nees F, Orfanos DP, Poustka L, Hohmann S, Holz N, Smolka MN, Vaidya N, Walter H, Whelan R, Schumann G, Lin X, Feng J, IMAGEN Consortium. Gene-environment interactions in the influence of maternal education on adolescent neurodevelopment using ABCD study. SCIENCE ADVANCES 2024; 10:eadp3751. [PMID: 39546599 PMCID: PMC11567010 DOI: 10.1126/sciadv.adp3751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024]
Abstract
Maternal education was strongly correlated with adolescent brain morphology, cognitive performances, and mental health. However, the molecular basis for the effects of maternal education on the structural neurodevelopment remains unknown. Here, we conducted gene-environment-wide interaction study using the Adolescent Brain Cognitive Development cohort. Seven genomic loci with significant gene-environment interactions (G×E) on regional gray matter volumes were identified, with enriched biological functions related to metabolic process, inflammatory process, and synaptic plasticity. Additionally, genetic overlapping results with behavioral and disease-related phenotypes indicated shared biological mechanism between maternal education modified neurodevelopment and related behavioral traits. Finally, by decomposing the multidimensional components of maternal education, we found that socioeconomic status, rather than family environment, played a more important role in modifying the genetic effects on neurodevelopment. In summary, our study provided analytical evidence for G×E effects regarding adolescent neurodevelopment and explored potential biological mechanisms as well as social mechanisms through which maternal education could modify the genetic effects on regional brain development.
Collapse
Affiliation(s)
- Runye Shi
- School of Data Science, Fudan University, Shanghai, China
| | - Xiao Chang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Gareth J. Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Arun L. W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, 68131 Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, 05405 Burlington, VT, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité–Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 “Trajectoires développementales en psychiatrie”, Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
| | | | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 “Trajectoires développementales en psychiatrie”, Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein Kiel University, Kiel, Germany
| | | | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Holz
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Michael N. Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité–Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | - Xiaolei Lin
- School of Data Science, Fudan University, Shanghai, China
| | - Jianfeng Feng
- School of Data Science, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, UK
| | | |
Collapse
|
22
|
Feng N, Mandal A, Jambhale A, Narnur P, Chen G, Akula N, Kramer R, Kolachana B, Xu Q, McMahon FJ, Lipska BK, Auluck PK, Marenco S. Schizophrenia risk-associated SNPs affect expression of microRNA 137 host gene: a postmortem study. Hum Mol Genet 2024; 33:1939-1947. [PMID: 39239979 PMCID: PMC12102068 DOI: 10.1093/hmg/ddae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
Common variants in the MicroRNA 137 host gene MIR137HG and its adjacent gene DPYD have been associated with schizophrenia risk and the latest Psychiatric Genomics Consortium (PGC). Genome-Wide Association Study on schizophrenia has confirmed and extended these findings. To elucidate the association of schizophrenia risk-associated SNPs in this genomic region, we examined the expression of both mature and immature transcripts of the miR-137 host gene (MIR137HG) in the dorsolateral prefrontal cortex (DLPFC) and subgenual anterior cingulate cortex (sgACC) of postmortem brain samples of donors with schizophrenia and psychiatrically-unaffected controls using qPCR and RNA-Seq approaches. No differential expression of miR-137, MIR137HG, or its transcripts was observed. Two schizophrenia risk-associated SNPs identified in the PGC study, rs11165917 (DLPFC: P = 2.0e-16; sgACC: P = 6.4e-10) and rs4274102 (DLPFC: P = 0.036; sgACC: P = 0.002), were associated with expression of the MIR137HG long non-coding RNA transcript MIR137HG-203 (ENST00000602672.2) in individuals of European ancestry. Carriers of the minor (risk) allele of rs11165917 had significantly lower expression of MIR137HG-203 compared with those carrying the major allele. However, we were unable to validate this result by short-read sequencing of RNA extracted from DLPFC or sgACC tissue. This finding suggests that immature transcripts of MIR137HG may contribute to genetic risk for schizophrenia.
Collapse
Affiliation(s)
- Ningping Feng
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Ajeet Mandal
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Ananya Jambhale
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Pranav Narnur
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Gang Chen
- Scientific and Statistical Computing Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, bldg 10, room 1D73, Bethesda, MD 20892, United States
| | - Nirmala Akula
- Human Genetics Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 35 Convent Dr. Bldg. 35, RM 1A202, MSC 3719, Bethesda, MD 20892, United States
| | - Robin Kramer
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Bhaskar Kolachana
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Qing Xu
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Francis J McMahon
- Human Genetics Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 35 Convent Dr. Bldg. 35, RM 1A202, MSC 3719, Bethesda, MD 20892, United States
| | - Barbara K Lipska
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Pavan K Auluck
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| | - Stefano Marenco
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 10 Center Drive, Bldg 10, room 4N218, Bethesda, MD 20892, United States
| |
Collapse
|
23
|
Savchenko RR, Skryabin NA. Transcription factor TCF4: structure, function, and associated diseases. Vavilovskii Zhurnal Genet Selektsii 2024; 28:770-779. [PMID: 39722673 PMCID: PMC11667571 DOI: 10.18699/vjgb-24-85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 12/28/2024] Open
Abstract
Our understanding of human genes - particularly their structure, functions, and regulatory mechanisms - is still limited. The biological role of approximately 20 % of human proteins has not been established yet, and the molecular functions of the known part of the proteome remain poorly understood. This hinders progress in basic and applied biological and medical sciences, especially in treating hereditary diseases, which are caused by mutations and polymorphic variants in individual genes. Therefore, it is crucial to comprehend the mechanisms of protein functioning to address this problem. This further emphasizes the importance of investigating gene functions and molecular pathogenetic pathways associated with single-gene inherited diseases. This review focuses on the TCF4 gene that encodes a transcription factor crucial for nervous system development and functioning. Pathogenic variants in this gene have been linked to a rare genetic disorder, Pitt-Hopkins syndrome, and TCF4 polymorphic variants are associated with several socially significant diseases, including various psychiatric disorders. The pathogenetic mechanisms of these conditions remain unexplored, and the knowledge about TCF4 upregulation and its target genes is limited. TCF4 can be expressed in various isoforms due to the complex structure and regulation of its gene, which complicates the investigation of the protein's functions. Here, we consider the structure and functions of the TCF4 transcription factor. We discuss its potential target genes and the possible loss-of-function pathogenetic mechanisms identified in animal and cellular models of Pitt-Hopkins syndrome. The review also examines the advantages and limitations of potential therapies for Pitt-Hopkins syndrome that are based on TCF4 dosage compensation or altering the activity of TCF4 target genes.
Collapse
Affiliation(s)
- R R Savchenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - N A Skryabin
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
24
|
Choudhary A, Peles D, Nayak R, Mizrahi L, Stern S. Current progress in understanding schizophrenia using genomics and pluripotent stem cells: A meta-analytical overview. Schizophr Res 2024; 273:24-38. [PMID: 36443183 DOI: 10.1016/j.schres.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/16/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022]
Abstract
Schizophrenia (SCZ) is a complex, heritable and polygenic neuropsychiatric disease, which disables the patients as well as decreases their life expectancy and quality of life. Common and rare variants studies on SCZ subjects have provided >100 genomic loci that hold importance in the context of SCZ pathophysiology. Transcriptomic studies from clinical samples have informed about the differentially expressed genes (DEGs) and non-coding RNAs in SCZ patients. Despite these advancements, no causative genes for SCZ were found and hence SCZ is difficult to recapitulate in animal models. In the last decade, induced Pluripotent Stem Cells (iPSCs)-based models have helped in understanding the neural phenotypes of SCZ by studying patient iPSC-derived 2D neuronal cultures and 3D brain organoids. Here, we have aimed to provide a simplistic overview of the current progress and advancements after synthesizing the enormous literature on SCZ genetics and SCZ iPSC-based models. Although further understanding of SCZ genetics and pathophysiological mechanisms using these technological advancements is required, the recent approaches have allowed to delineate important cellular mechanisms and biological pathways affected in SCZ.
Collapse
Affiliation(s)
- Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
25
|
Muhtaseb AW, Duan J. Modeling common and rare genetic risk factors of neuropsychiatric disorders in human induced pluripotent stem cells. Schizophr Res 2024; 273:39-61. [PMID: 35459617 PMCID: PMC9735430 DOI: 10.1016/j.schres.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Recent genome-wide association studies (GWAS) and whole-exome sequencing of neuropsychiatric disorders, especially schizophrenia, have identified a plethora of common and rare disease risk variants/genes. Translating the mounting human genetic discoveries into novel disease biology and more tailored clinical treatments is tied to our ability to causally connect genetic risk variants to molecular and cellular phenotypes. When combined with the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) nuclease-mediated genome editing system, human induced pluripotent stem cell (hiPSC)-derived neural cultures (both 2D and 3D organoids) provide a promising tractable cellular model for bridging the gap between genetic findings and disease biology. In this review, we first conceptualize the advances in understanding the disease polygenicity and convergence from the past decade of iPSC modeling of different types of genetic risk factors of neuropsychiatric disorders. We then discuss the major cell types and cellular phenotypes that are most relevant to neuropsychiatric disorders in iPSC modeling. Finally, we critically review the limitations of iPSC modeling of neuropsychiatric disorders and outline the need for implementing and developing novel methods to scale up the number of iPSC lines and disease risk variants in a systematic manner. Sufficiently scaled-up iPSC modeling and a better functional interpretation of genetic risk variants, in combination with cutting-edge CRISPR/Cas9 gene editing and single-cell multi-omics methods, will enable the field to identify the specific and convergent molecular and cellular phenotypes in precision for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdurrahman W Muhtaseb
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
26
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
27
|
Gober R, Dallmeier J, Davis D, Brzostowicki D, de Rivero Vaccari JP, Cyr B, Barreda A, Sun X, Gultekin SH, Garamszegi S, Scott W, Vontell R. Increased inflammasome protein expression identified in microglia from postmortem brains with schizophrenia. J Neuropathol Exp Neurol 2024; 83:951-966. [PMID: 38904417 PMCID: PMC11487111 DOI: 10.1093/jnen/nlae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder that involves an inflammatory response thought to be characterized by microglial activation. The inflammasome complex may play critical roles in the pathomechanism of neuroinflammation but how this relates to SCZ remains unclear. In this study, we performed an immunohistochemical (IHC) analysis to compare the expression of inflammasome proteins in brain tissue from donors with SCZ (n = 16) and non-psychiatric donors (NP; n = 13) isolated from the superior frontal cortex (SFC), superior temporal cortex, and anterior cingulate cortex brain regions. To assess changes in the cell populations that express key inflammasome proteins, we performed IHC analyses of apoptosis-associated speck-like protein containing a CARD (ASC), nod-like receptor protein 3 (NLRP3), and interleukin (IL)-18 to determine if these proteins are expressed in microglia, astrocytes, oligodendrocytes, or neurons. Inflammasome proteins were expressed mainly in microglia from SCZ and NP brains. Increased numbers of microglia were present in the SFC of SCZ brains and exhibited higher inflammasome protein expression of ASC, NLRP3, and IL-18 compared to NPs. These findings suggest that increased inflammasome signaling may contribute to the pathology underlying SCZ.
Collapse
Affiliation(s)
- Ryan Gober
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Julian Dallmeier
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - David Davis
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Daniel Brzostowicki
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami, Miami, FL, United States
| | - Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami, Miami, FL, United States
| | - Ayled Barreda
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Xiaoyan Sun
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sakir Humayun Gultekin
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Susanna Garamszegi
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - William Scott
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Regina Vontell
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
28
|
Khoodoruth MAS, Khoodoruth WNCK, Uroos M, Al-Abdulla M, Khan YS, Mohammad F. Diagnostic and mechanistic roles of MicroRNAs in neurodevelopmental & neurodegenerative disorders. Neurobiol Dis 2024; 202:106717. [PMID: 39461569 DOI: 10.1016/j.nbd.2024.106717] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 09/15/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024] Open
Abstract
MicroRNAs (miRNAs) are emerging as crucial elements in the regulation of gene expression, playing a significant role in the underlying neurobiology of a wide range of neuropsychiatric disorders. This review examines the intricate involvement of miRNAs in neuropsychiatric disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Fragile X syndrome (FXS), autism spectrum disorder (ASD), attention-deficit hyperactivity disorder (ADHD), Tourette syndrome (TS), schizophrenia (SCZ), and mood disorders. This review highlights how miRNA dysregulation can illuminate the molecular pathways of these diseases and potentially serve as biomarkers for early diagnosis and prognosis. Specifically, miRNAs' ability to target genes critical to the pathology of neurodegenerative diseases, their role in the development of trinucleotide repeat and neurodevelopmental disorders, and their distinctive patterns in SCZ and mood disorders are discussed. The review also stresses the value of miRNAs in precision neuropsychiatry, where they could predict treatment outcomes and aid in disease management. Furthermore, the study of conserved miRNAs in model organisms like Drosophila underscores their broad utility and provides deeper mechanistic insights into their biological functions. This comprehensive examination of miRNAs across various conditions advocates for their integration into clinical practice, promising advancements in personalized healthcare for neurological and psychiatric conditions.
Collapse
Affiliation(s)
- Mohamed Adil Shah Khoodoruth
- Child and Adolescent Mental Health Service, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar
| | | | | | - Majid Al-Abdulla
- Mental Health Service, Hamad Medical Corporation, Doha, Qatar; College of Medicine, Qatar University, Doha, Qatar
| | - Yasser Saeed Khan
- Child and Adolescent Mental Health Service, Hamad Medical Corporation, Doha, Qatar
| | - Farhan Mohammad
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Qatar.
| |
Collapse
|
29
|
Lorincz-Comi N, Yang Y, Ajayakumar J, Mews M, Bermudez V, Bush W, Zhu X. HORNET: Tools to find genes with causal evidence and their regulatory networks using eQTLs. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.28.24316273. [PMID: 39574873 PMCID: PMC11581076 DOI: 10.1101/2024.10.28.24316273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Motivation Nearly two decades of genome-wide association studies (GWAS) have identify thousands of disease-associated genetic variants, but very few genes with evidence of causality. Recent methodological advances demonstrate that Mendelian Randomization (MR) using expression quantitative loci (eQTLs) as instrumental variables can detect potential causal genes. However, existing MR approaches are not well suited to handle the complexity of eQTL GWAS data structure and so they are subject to bias, inflation, and incorrect inference. Results We present a whole-genome regulatory network analysis tool (HORNET), which is a comprehensive set of statistical and computational tools to perform genome-wide searches for causal genes using summary level GWAS data that is robust to biases from multiple sources. Applying HORNET to schizophrenia, we identified differential magnitudes of gene expression causality. Applying HORNET to schizophrenia, we identified differential magnitudes of gene expression causality across different brain tissues. Availability and Implementation Freely available at https://github.com/noahlorinczcomi/HORNETor Mac, Windows, and Linux users. Contact njl96@case.edu .
Collapse
|
30
|
Funato Y, Mimura M, Nunomura K, Lin B, Fujii S, Haruta J, Miki H. Development of a high-throughput screening system targeting the protein-protein interactions between PRL and CNNM. Sci Rep 2024; 14:25432. [PMID: 39455715 PMCID: PMC11511866 DOI: 10.1038/s41598-024-76269-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Phosphatase of regenerating liver (PRL) is an oncogenic protein that promotes tumor progression by directly binding to cyclin M (CNNM) membrane proteins and inhibiting their Mg2+ efflux activity. In this study, we have developed a high-throughput screening system to detect the interactions between PRL and CNNM proteins based on homogenous time-resolved fluorescence resonance energy transfer (HTR-FRET, HTRF). We optimized the tag sequences attached to the recombinant proteins of the CNNM4 CBS domains and PRL3 lacking the carboxyl terminal CAAX motif, and successfully detected the interaction by observing the FRET signal in the mixture of the tagged proteins and fluorophore-conjugated antibodies. Moreover, we performed compound library screening using this system and discovered several compounds that could efficiently inhibit the PRL-CNNM interaction. Characterization of one candidate compound revealed that it was relatively stable compared with thienopyridone, a known inhibitor of the PRL-CNNM interaction. The candidate compound can also inhibit PRL function in cells: suppression of CNNM-dependent Mg2+ efflux, and has sufficient in vitro drug metabolism and pharmacokinetic properties. Overall, these results demonstrate the effectiveness of this screening system for identifying novel inhibitors of the PRL-CNNM interaction, which could contribute to the development of novel anti-cancer drugs.
Collapse
Affiliation(s)
- Yosuke Funato
- Laboratory of Biorecognition Chemistry, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan.
| | - Mai Mimura
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Kazuto Nunomura
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Bangzhong Lin
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Shintarou Fujii
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Junichi Haruta
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Hiroaki Miki
- Laboratory of Biorecognition Chemistry, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan.
| |
Collapse
|
31
|
Burette AC, Vihma H, Smith AL, Ozarkar SS, Bennett J, Amaral DG, Philpot BD. Transcription factor 4 expression in the developing non-human primate brain: a comparative analysis with the mouse brain. Front Neuroanat 2024; 18:1478689. [PMID: 39502395 PMCID: PMC11534587 DOI: 10.3389/fnana.2024.1478689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
Transcription factor 4 (TCF4) has been implicated in a range of neuropsychiatric disorders, including major depressive disorder, bipolar disorder, and schizophrenia. Mutations or deletions in TCF4 cause Pitt-Hopkins syndrome (PTHS), a rare neurodevelopmental disorder. A detailed understanding of its spatial expression across the developing brain is necessary for comprehending TCF4 biology and, by extension, to develop effective treatments for TCF4-associated disorders. However, most current knowledge is derived from mouse models, which are invaluable for preclinical studies but may not fully capture the complexities of human neuropsychiatric phenotypes. This study compared TCF4 expression in the developing mouse brain to its regional and cellular expression patterns in normal prenatal, neonatal, and young adult rhesus macaque brains, a species more relevant to human neurodevelopment. While the general developmental expression of TCF4 is largely conserved between macaques and mice, we saw several interspecies differences. Most notably, a distinct layered pattern of TCF4 expression was clear in the developing macaque neocortex but largely absent in the mouse brain. High TCF4 expression was seen in the inner dentate gyrus of adult mice but not in macaques. Conversely, TCF4 expression was higher in the adult macaque striatum compared to the mouse striatum. Further research is needed to show the significance of these interspecies differences. Still, they underscore the importance of integrating rodent and primate studies to comprehensively understand TCF4 function and its implications for human disorders. Moreover, the primate-specific expression patterns of TCF4 will inform genetic and other therapeutic strategies to treat TCF4-associated disorders.
Collapse
Affiliation(s)
- Alain C. Burette
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Hanna Vihma
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Audrey L. Smith
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Siddhi S. Ozarkar
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jeff Bennett
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Davis, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - David G. Amaral
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Davis, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Benjamin D. Philpot
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
32
|
Sullivan KA, Lane M, Cashman M, Miller JI, Pavicic M, Walker AM, Cliff A, Romero J, Qin X, Mullins N, Docherty A, Coon H, Ruderfer DM, Garvin MR, Pestian JP, Ashley-Koch AE, Beckham JC, McMahon B, Oslin DW, Kimbrel NA, Jacobson DA, Kainer D. Analyses of GWAS signal using GRIN identify additional genes contributing to suicidal behavior. Commun Biol 2024; 7:1360. [PMID: 39433874 PMCID: PMC11494055 DOI: 10.1038/s42003-024-06943-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Genome-wide association studies (GWAS) identify genetic variants underlying complex traits but are limited by stringent genome-wide significance thresholds. We present GRIN (Gene set Refinement through Interacting Networks), which increases confidence in the expanded gene set by retaining genes strongly connected by biological networks when GWAS thresholds are relaxed. GRIN was validated on both simulated interrelated gene sets as well as multiple GWAS traits. From multiple GWAS summary statistics of suicide attempt, a complex phenotype, GRIN identified additional genes that replicated across independent cohorts and retained biologically interrelated genes despite a relaxed significance threshold. We present a conceptual model of how these retained genes interact through neurobiological pathways that may influence suicidal behavior, and identify existing drugs associated with these pathways that would not have been identified under traditional GWAS thresholds. We demonstrate GRIN's utility in boosting GWAS results by increasing the number of true positive genes identified from GWAS results.
Collapse
Affiliation(s)
- Kyle A Sullivan
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Matthew Lane
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Mikaela Cashman
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory Berkeley, California, CA, USA
| | - J Izaak Miller
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Mirko Pavicic
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Angelica M Walker
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Ashley Cliff
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Jonathon Romero
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Xuejun Qin
- Durham Veterans Affairs Health Care System, Durham, NC, USA
- Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Niamh Mullins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna Docherty
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hilary Coon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
- Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Douglas M Ruderfer
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael R Garvin
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - John P Pestian
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Allison E Ashley-Koch
- Duke University School of Medicine, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Jean C Beckham
- Durham Veterans Affairs Health Care System, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
- VISN 6 Mid-Atlantic Mental Illness Research, Durham Veterans Affairs Health Care System, Durham, NC, USA
| | - Benjamin McMahon
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - David W Oslin
- VISN 4 Mental Illness Research, Education, and Clinical Center, Center of Excellence, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan A Kimbrel
- Durham Veterans Affairs Health Care System, Durham, NC, USA.
- Duke University School of Medicine, Duke University, Durham, NC, USA.
- VISN 6 Mid-Atlantic Mental Illness Research, Durham Veterans Affairs Health Care System, Durham, NC, USA.
- VA Health Services Research and Development Center of Innovation to Accelerate Discovery and Practice Transformation, Durham, NC, USA.
| | - Daniel A Jacobson
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA.
| | - David Kainer
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA.
- Centre of Excellence for Plant Success in Nature and Agriculture, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
33
|
Heider J, González EP, Hartmann SM, Kannaiyan N, Vogel S, Wüst R, Fallgatter AJ, Rossner MJ, Kraushaar U, Volkmer H. Aberrant neuronal connectivity and network activity of neurons derived from patients with idiopathic schizophrenia. Neurobiol Dis 2024; 201:106678. [PMID: 39307399 DOI: 10.1016/j.nbd.2024.106678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder with a strong genetic determinant. A major hypothesis to explain disease aetiology comprises synaptic dysfunction associated with excitatory-inhibitory imbalance of synaptic transmission, ultimately contributing to impaired network oscillation and cognitive deficits associated with the disease. Here, we studied the morphological and functional properties of a highly defined co-culture of GABAergic and glutamatergic neurons derived from induced pluripotent stem cells (iPSC) from patients with idiopathic SCZ. Our results indicate upregulation of synaptic genes and increased excitatory synapse formation on GABAergic neurons in co-cultures. In parallel, we observed decreased lengths of axon initial segments, concordant with data from postmortem brains from patients with SCZ. In line with increased synapse density, patch-clamp analyses revealed markedly increased spontaneous excitatory postsynaptic currents (EPSC) recorded from GABAergic SCZ neurons. Finally, MEA recordings from neuronal networks indicate increased strength of network activity, potentially in response to altered synaptic transmission and E-I balance in the co-cultures. In conclusion, our results suggest selective deregulation of neuronal activity in SCZ samples, providing evidence for altered synapse formation and synaptic transmission as a potential base for aberrant network synchronization.
Collapse
Affiliation(s)
- Johanna Heider
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Emilio Pardo González
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany; International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Sophia-Marie Hartmann
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany; International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Nirmal Kannaiyan
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Sabrina Vogel
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Richard Wüst
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany; German Center for Mental Health (DZPG), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Andreas J Fallgatter
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany; German Center for Mental Health (DZPG), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Moritz J Rossner
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany
| | - Hansjürgen Volkmer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany; International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany; German Center for Mental Health (DZPG), Partner Site Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
34
|
Zhang H, McCarroll A, Peyton L, Díaz de León-Guerrerro S, Zhang S, Gowda P, Sirkin D, ElAchwah M, Duhe A, Wood WG, Jamison B, Tracy G, Pollak R, Hart RP, Pato CN, Mulle JG, Sanders AR, Pang ZP, Duan J. Scaled and efficient derivation of loss-of-function alleles in risk genes for neurodevelopmental and psychiatric disorders in human iPSCs. Stem Cell Reports 2024; 19:1489-1504. [PMID: 39270650 PMCID: PMC11561461 DOI: 10.1016/j.stemcr.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/08/2024] [Accepted: 08/10/2024] [Indexed: 09/15/2024] Open
Abstract
Translating genetic findings for neurodevelopmental and psychiatric disorders (NPDs) into actionable disease biology would benefit from large-scale and unbiased functional studies of NPD genes. Leveraging the cytosine base editing (CBE) system, we developed a pipeline for clonal loss-of-function (LoF) allele mutagenesis in human induced pluripotent stem cells (hiPSCs) by introducing premature stop codons (iSTOP) that lead to mRNA nonsense-mediated decay (NMD) or protein truncation. We tested the pipeline for 23 NPD genes on 3 hiPSC lines and achieved highly reproducible, efficient iSTOP editing in 22 genes. Using RNA sequencing (RNA-seq), we confirmed their pluripotency, absence of chromosomal abnormalities, and NMD. Despite high editing efficiency, three schizophrenia risk genes (SETD1A, TRIO, and CUL1) only had heterozygous LoF alleles, suggesting their essential roles for cell growth. We found that CUL1-LoF reduced neurite branches and synaptic puncta density. This iSTOP pipeline enables a scaled and efficient LoF mutagenesis of NPD genes, yielding an invaluable shareable resource.
Collapse
Affiliation(s)
- Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Ada McCarroll
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Lilia Peyton
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Sol Díaz de León-Guerrerro
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL, USA
| | - Prarthana Gowda
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - David Sirkin
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Mahmoud ElAchwah
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Alexandra Duhe
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Whitney G Wood
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Brandon Jamison
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Gregory Tracy
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Rebecca Pollak
- Center for Advanced Biotechnology and Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Carlos N Pato
- Center for Advanced Biotechnology and Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Jennifer G Mulle
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Advanced Biotechnology and Medicine, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA; Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL, USA; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
35
|
Mazzarotto F, Monteleone P, Minelli A, Mattevi S, Cascino G, Rocca P, Rossi A, Bertolino A, Aguglia E, Altamura C, Amore M, Bellomo A, Bucci P, Collantoni E, Dell'Osso L, Di Fabio F, Fagiolini A, Giuliani L, Marchesi C, Martinotti G, Montemagni C, Pinna F, Pompili M, Rampino A, Roncone R, Siracusano A, Vita A, Zeppegno P, Galderisi S, Gennarelli M, Maj M. Genetic determinants of coping, resilience and self-esteem in schizophrenia suggest a primary role for social factors and hippocampal neurogenesis. Psychiatry Res 2024; 340:116107. [PMID: 39096746 DOI: 10.1016/j.psychres.2024.116107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Schizophrenia is a severe psychiatric disorder, associated with a reduction in life expectancy of 15-20 years. Available treatments are at least partially effective in most affected individuals, and personal resources such as resilience (successful adaptation despite adversity) and coping abilities (strategies used to deal with stressful or threatening situations), are important determinants of disease outcomes and long-term sustained recovery. Published findings support the existence of a genetic background underlying resilience and coping, with variable heritability estimates. However, genome-wide analyses concerning the genetic determinants of these personal resources, especially in the context of schizophrenia, are lacking. Here, we performed a genome-wide association study coupled with accessory analyses to investigate potential genetic determinants of resilience, coping and self-esteem in 490 schizophrenia patients. Results revealed a complex genetic background partly overlapping with that of neuroticism, worry and schizophrenia itself and support the importance of social aspects in shapingthese psychological constructs. Hippocampal neurogenesis and lipid metabolism appear to be potentially relevant biological underpinnings, and specific miRNAs such as miR-124 and miR-137 may warrant further studies as potential biomarkers. In conclusion, this study represents an important first step in the identification of genetic and biological correlates shaping resilience, coping resources and self-esteem in schizophrenia.
Collapse
Affiliation(s)
- Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Palmiero Monteleone
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Stefania Mattevi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giammarco Cascino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Paola Rocca
- Department of Neuroscience, Section of Psychiatry, University of Turin, Turin, Italy
| | - Alessandro Rossi
- Section of Psychiatry, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Bertolino
- Department of Neurological and Psychiatric Sciences, University of Bari, Bari, Italy
| | - Eugenio Aguglia
- Department of Clinical and Molecular Biomedicine, Psychiatry Unit, University of Catania, Catania, Italy
| | - Carlo Altamura
- Department of Psychiatry, University of Milan, Milan, Italy
| | - Mario Amore
- Section of Psychiatry, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Antonello Bellomo
- Psychiatry Unit, Department of Medical Sciences, University of Foggia, Foggia, Italy
| | - Paola Bucci
- Department of Psychiatry, University of Campania "Luigi Vanvitelli" Naples, Italy
| | - Enrico Collantoni
- Psychiatric Clinic, Department of Neurosciences, University of Padua, Padua, Italy
| | - Liliana Dell'Osso
- Section of Psychiatry, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fabio Di Fabio
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | - Andrea Fagiolini
- Department of Molecular Medicine and Clinical Department of Mental Health, University of Siena, Siena, Italy
| | - Luigi Giuliani
- Department of Psychiatry, University of Campania "Luigi Vanvitelli" Naples, Italy
| | - Carlo Marchesi
- Department of Neuroscience, Psychiatry Unit, University of Parma, Parma, Italy
| | - Giovanni Martinotti
- Department of Neuroscience and Imaging, G. D'Annunzio University, Chieti, Italy
| | - Cristiana Montemagni
- Department of Neuroscience, Section of Psychiatry, University of Turin, Turin, Italy
| | - Federica Pinna
- Section of Psychiatry, Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Cagliari, Italy
| | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, S. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Antonio Rampino
- Department of Neurological and Psychiatric Sciences, University of Bari, Bari, Italy
| | - Rita Roncone
- Unit of Psychiatry, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alberto Siracusano
- Department of Systems Medicine, Psychiatry and Clinical Psychology Unit, Tor Vergata University of Rome, Rome, Italy
| | - Antonio Vita
- Psychiatric Unit, School of Medicine, University of Brescia, Brescia, Italy; Department of Mental Health, Spedali Civili Hospital, Brescia, Italy
| | - Patrizia Zeppegno
- Department of Translational Medicine, Psychiatric Unit, University of Eastern Piedmont, Novara, Italy
| | - Silvana Galderisi
- Department of Psychiatry, University of Campania "Luigi Vanvitelli" Naples, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Mario Maj
- Department of Psychiatry, University of Campania "Luigi Vanvitelli" Naples, Italy
| |
Collapse
|
36
|
Hughes H, Brady LJ, Schoonover KE. GABAergic dysfunction in postmortem dorsolateral prefrontal cortex: implications for cognitive deficits in schizophrenia and affective disorders. Front Cell Neurosci 2024; 18:1440834. [PMID: 39381500 PMCID: PMC11458443 DOI: 10.3389/fncel.2024.1440834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
The microcircuitry within superficial layers of the dorsolateral prefrontal cortex (DLPFC), composed of excitatory pyramidal neurons and inhibitory GABAergic interneurons, has been suggested as the neural substrate of working memory performance. In schizophrenia, working memory impairments are thought to result from alterations of microcircuitry within the DLPFC. GABAergic interneurons, in particular, are crucially involved in synchronizing neural activity at gamma frequency, the power of which increases with working memory load. Alterations of GABAergic interneurons, particularly parvalbumin (PV) and somatostatin (SST) subtypes, are frequently observed in schizophrenia. Abnormalities of GABAergic neurotransmission, such as deficiencies in the 67 kDA isoform of GABA synthesis enzyme (GAD67), vesicular GABA transporter (vGAT), and GABA reuptake transporter 1 (GAT1) in presynaptic boutons, as well as postsynaptic alterations in GABA A receptor subunits further contribute to impaired inhibition. This review explores GABAergic abnormalities of the postmortem DLPFC in schizophrenia, with a focus on the roles of interneuron subtypes involved in cognition, and GABAergic neurotransmission within presynaptic boutons and postsynaptic alterations. Where available, comparisons between schizophrenia and affective disorders that share cognitive pathology such as bipolar disorder and major depressive disorder will be made. Challenges in directly measuring GABA levels are addressed, emphasizing the need for innovative techniques. Understanding GABAergic abnormalities and their implications for neural circuit dysfunction in schizophrenia is crucial for developing targeted therapies.
Collapse
Affiliation(s)
- Hannah Hughes
- Graduate Biomedical Sciences Program, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
| | - Lillian J. Brady
- Department of Psychiatry, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Tuskegee, AL, United States
| | - Kirsten E. Schoonover
- Department of Psychiatry, School of Medicine, University of Alabama at Birmingham, Tuskegee, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Tuskegee, AL, United States
- Department of Psychology and Sociology, College of Arts and Sciences, Tuskegee University, Tuskegee, AL, United States
| |
Collapse
|
37
|
Yang Z, Yao S, Xu Y, Zhang X, Shi Y, Wang L, Cui D. Identification of a Predictive Model for Schizophrenia Based on SNPs in a Chinese Population. Neuropsychiatr Dis Treat 2024; 20:1553-1561. [PMID: 39139656 PMCID: PMC11321330 DOI: 10.2147/ndt.s466554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Background Schizophrenia is a devastating mental disease with high heritability. A growing number of susceptibility genes associated with schizophrenia, as well as their corresponding SNPs loci, have been revealed by genome-wide association studies. However, using SNPs as predictors of disease and diagnosis remains difficult. Here, we aimed to uncover susceptibility SNPs in a Chinese population and to construct a prediction model for schizophrenia. Methods A total of 210 participants, including 70 patients with schizophrenia, 70 patients with bipolar disorder, and 70 healthy controls, were enrolled in this study. We estimated 14 SNPs using published risk loci of schizophrenia, and used these SNPs to build a model for predicting schizophrenia via comparison of genotype frequencies and regression. We evaluated the efficacy of the diagnostic model in schizophrenia and control patients using ROC curves and then used the 70 patients with bipolar disorder to evaluate the model's differential diagnostic efficacy. Results 5 SNPs were selected to construct the model: rs148415900, rs71428218, rs4666990, rs112222723 and rs1716180. Correlation analysis results suggested that, compared with the risk SNP of 0, the risk SNP of 3 was associated with an increased risk of schizophrenia (OR = 13.00, 95% CI: 2.35-71.84, p = 0.003). The ROC-AUC of this prediction model for schizophrenia was 0.719 (95% CI: 0.634-0.804), with the greatest sensitivity and specificity being 60% and 80%, respectively. The ROC-AUC of the model in distinguishing between schizophrenia and bipolar disorder was 0.591 (95% CI: 0.497-0.686), with the greatest sensitivity and specificity being 60% and 55.7%, respectively. Conclusion The SNP risk score prediction model had good performance in predicting schizophrenia. To the best of our knowledge, previous studies have not applied SNP-based models to differentiate between cases of schizophrenia and other mental illnesses. It could have several potential clinical applications, including shaping disease diagnosis, treatment, and outcomes.
Collapse
Affiliation(s)
- Zhiying Yang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Shun Yao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yichong Xu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaoqing Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuan Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Lijun Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
38
|
Parkins EV, Gross C. Small Differences and Big Changes: The Many Variables of MicroRNA Expression and Function in the Brain. J Neurosci 2024; 44:e0365242024. [PMID: 39111834 PMCID: PMC11308354 DOI: 10.1523/jneurosci.0365-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 08/10/2024] Open
Abstract
MicroRNAs are emerging as crucial regulators within the complex, dynamic environment of the synapse, and they offer a promising new avenue for the treatment of neurological disease. These small noncoding RNAs modify gene expression in several ways, including posttranscriptional modulation via binding to complementary and semicomplementary sites on target mRNAs. This rapid, finely tuned regulation of gene expression is essential to meet the dynamic demands of the synapse. Here, we provide a detailed review of the multifaceted world of synaptic microRNA regulation. We discuss the many mechanisms by which microRNAs regulate gene expression at the synapse, particularly in the context of neuronal plasticity. We also describe the various factors, such as age, sex, and neurological disease, that can influence microRNA expression and activity in neurons. In summary, microRNAs play a crucial role in the intricate and quickly changing functional requirements of the synapse, and context is essential in the study of microRNAs and their potential therapeutic applications.
Collapse
Affiliation(s)
- Emma V Parkins
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, Ohio 45229
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Christina Gross
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, Ohio 45229
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229
| |
Collapse
|
39
|
Kong L, Chen Y, Shen Y, Zhang D, Wei C, Lai J, Hu S. Progress and Implications from Genetic Studies of Bipolar Disorder. Neurosci Bull 2024; 40:1160-1172. [PMID: 38206551 PMCID: PMC11306703 DOI: 10.1007/s12264-023-01169-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 01/12/2024] Open
Abstract
With the advancements in gene sequencing technologies, including genome-wide association studies, polygenetic risk scores, and high-throughput sequencing, there has been a tremendous advantage in mapping a detailed blueprint for the genetic model of bipolar disorder (BD). To date, intriguing genetic clues have been identified to explain the development of BD, as well as the genetic association that might be applied for the development of susceptibility prediction and pharmacogenetic intervention. Risk genes of BD, such as CACNA1C, ANK3, TRANK1, and CLOCK, have been found to be involved in various pathophysiological processes correlated with BD. Although the specific roles of these genes have yet to be determined, genetic research on BD will help improve the prevention, therapeutics, and prognosis in clinical practice. The latest preclinical and clinical studies, and reviews of the genetics of BD, are analyzed in this review, aiming to summarize the progress in this intriguing field and to provide perspectives for individualized, precise, and effective clinical practice.
Collapse
Affiliation(s)
- Lingzhuo Kong
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yiqing Chen
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuting Shen
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Danhua Zhang
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Wei
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jianbo Lai
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Shaohua Hu
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, 310003, China.
- Brain Research Institute of Zhejiang University, Hangzhou, 310003, China.
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, 310003, China.
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
40
|
Usta Saglam NG, Duz MB, Salman Yilmaz S, Ozen M, Balcioglu I. Comparison of microRNA expression levels in patients with schizophrenia before and after electroconvulsive therapy. Psychiatr Genet 2024; 34:79-85. [PMID: 38842000 DOI: 10.1097/ypg.0000000000000371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
OBJECTIVE Exploring the role of microRNAs in the antipsychotic efficacy of electroconvulsive therapy (ECT) will contribute to understanding the underlying mechanism through which ECT exerts its therapeutic effects. The primary objective of this study was to identify microRNA alterations before and after ECT in patients with schizophrenia. METHODS We compared microarray-based microRNA profiles in peripheral blood from eight patients with schizophrenia before and after ECT and eight healthy controls. Then, we aimed to validate selected differentially expressed microRNAs in 30 patients with schizophrenia following a course of ECT, alongside 30 healthy controls by using quantitative reverse-transcription PCR. RESULTS Microarray-based expression profiling revealed alterations in 681 microRNAs when comparing pre- and post-ECT samples. Subsequent quantitative reverse-transcription PCR analysis of the selected microRNAs (miR-20a-5p and miR-598) did not reveal any statistical differences between pre- and post-ECT samples nor between pre-ECT samples and those of healthy controls. CONCLUSION As neuroepigenetic studies on ECT are still in their infancy, the results reported in this study are best interpreted as exploratory outcomes. Additional studies are required to explore the potential epigenetic mechanisms underlying the therapeutic efficacy of ECT.
Collapse
Affiliation(s)
| | - Mehmet Bugrahan Duz
- Department of Medical Genetics, Istanbul Memorial Hospital
- Department of Medical Genetics, Istanbul Arel University
| | - Seda Salman Yilmaz
- Department of Medical Genetics, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa
- Department of Medical Services and Techniques Medical Monitoring Techniques Pr., Vocational School of Health Services, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Mustafa Ozen
- Department of Medical Genetics, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
41
|
Xavier G, Mauer J, Ota VK, Santoro ML, Belangero SI. Influence of antipsychotic drugs on microRNA expression in schizophrenia patients - A systematic review. J Psychiatr Res 2024; 176:163-172. [PMID: 38870782 DOI: 10.1016/j.jpsychires.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/23/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Schizophrenia (SCZ) is a severe psychiatric disorder with unclear pathophysiology. Moreover, there is no specific biological marker to help clinicians to define a diagnosis, and medication is decided according to the psychiatrist's experience. In this scenario, microRNAs (miRNAs), which are small noncoding RNA molecules that regulate several genes, emerge as potential peripheral biomarkers to help not only the evaluation of the disease state but also the treatment response. Here, we systematically reviewed indexed literature and evaluated follow-up studies investigating the changes in miRNA expression due to antipsychotic treatment. We also assessed target genes and performed pathway enrichment analysis of miRNAs listed in this systematic review. A total of 11 studies were selected according to research criteria, and we observed that 28 miRNAs play a relevant role in schizophrenia pathogenesis or response to antipsychotic treatment, seven of those of extreme interest as possible biomarkers either for condition or treatment. Predicted targets of the miRNAs reviewed here were previously associated with schizophrenia in genome-wide studies, and pathway analysis showed enrichment for genes related to neural processes. With this review, we expect to highlight the importance of miRNAs in schizophrenia pathogenesis and its treatment and point out interesting miRNAs to future studies.
Collapse
Affiliation(s)
- Gabriela Xavier
- LiNC, Laboratory of Integrative Neuroscience - Department of Psychiatry - Universidade Federal de São Paulo, Brazil; Genetics Division - Department of Morphology and Genetics - Universidade Federal de São Paulo, Brazil
| | - Jessica Mauer
- LiNC, Laboratory of Integrative Neuroscience - Department of Psychiatry - Universidade Federal de São Paulo, Brazil; Genetics Division - Department of Morphology and Genetics - Universidade Federal de São Paulo, Brazil
| | - Vanessa K Ota
- LiNC, Laboratory of Integrative Neuroscience - Department of Psychiatry - Universidade Federal de São Paulo, Brazil; Genetics Division - Department of Morphology and Genetics - Universidade Federal de São Paulo, Brazil
| | - Marcos L Santoro
- LiNC, Laboratory of Integrative Neuroscience - Department of Psychiatry - Universidade Federal de São Paulo, Brazil; Disciplina de Biologia Molecular - Departamento de Bioquímica - Universidade Federal de São Paulo, Brazil
| | - Sintia I Belangero
- LiNC, Laboratory of Integrative Neuroscience - Department of Psychiatry - Universidade Federal de São Paulo, Brazil; Genetics Division - Department of Morphology and Genetics - Universidade Federal de São Paulo, Brazil.
| |
Collapse
|
42
|
Mihoub O, Ben Chaaben A, Boukouaci W, Lajnef M, Ayari F, El Kefi H, Ben Ammar H, Abazza H, El Hechmi Z, Guemira F, Leboyer M, Tamouza R, Kharrat M. CSMD1 rs10503253 increases schizophrenia risk in a Tunisian population-group. L'ENCEPHALE 2024; 50:380-385. [PMID: 37748985 DOI: 10.1016/j.encep.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 09/27/2023]
Abstract
OBJECTIVES Schizophrenia is a complex and chronic neuropsychiatric disorder. Recent genome-wide association studies have identified several at risk genetic variants, including two single nucleotide polymorphisms, namely the rs10503253 and the rs1270942 respectively located in the CSMD1 and the CFB loci. The present case-control study was designed to assess potential associations between the two variants and the risk of developing schizophrenia and disease severity. Further we demonstrate the relationship between these variants and clinical characteristics in a population-group from Tunisia. PATIENTS AND METHODS In total, 216 patients diagnosed with schizophrenia along with176 healthy controls were included in this case-control study. The molecular analysis of the two polymorphisms was performed using tetra the Primer Amplification Refractory Mutation System-Polymerase Chain method. The statistical analysis was done using Compare V2.1 software, and correlations between genetic results and clinical characteristics were examined by Kruskal-Wallis testing. RESULTS The frequency of the rs10503253A allele was found significantly higher among patients with schizophrenia as compared to healthy controls and associated with high negative PANSS scores. While no association was found concerning the implication of the rs1270942 variant in schizophrenia risk, a positive correlation with high positive PANSS scores was further observed. CONCLUSION The present finding confirms the previously reported association between the Cub and Sushi multiple Domain 1 rs10503253A allele and the risk to develop schizophrenia and identified the rs1270942 variant as a potential disease risk modifier. Such observations may be important for the definition of the susceptible immunogenetic background in North African individuals at risk to develop mental disorders.
Collapse
Affiliation(s)
- Ons Mihoub
- Laboratory of Human Genetics (LR99ES10), Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia; Inserm U955 IMRB, Translational Neuropsychiatry Laboratory and Paris-Est Créteil University, 94010 Créteil, France.
| | - Arij Ben Chaaben
- Laboratory of Human Genetics (LR99ES10), Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Wahid Boukouaci
- Inserm U955 IMRB, Translational Neuropsychiatry Laboratory and Paris-Est Créteil University, 94010 Créteil, France
| | - Mohamed Lajnef
- Inserm U955 IMRB, Translational Neuropsychiatry Laboratory and Paris-Est Créteil University, 94010 Créteil, France
| | - Fayza Ayari
- Clinical Biology Department, Salah Azaiz Institute, Tunis, Tunisia
| | - Hamdi El Kefi
- Department of Psychiatry, Military Hospital of Tunis, Tunis, Tunisia
| | - Hanen Ben Ammar
- Department of Psychiatry F, Razi Hospital, Mannouba, Tunisia
| | - Hajer Abazza
- Laboratory of Human Genetics (LR99ES10), Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | | | - Fathi Guemira
- Clinical Biology Department, Salah Azaiz Institute, Tunis, Tunisia
| | - Marion Leboyer
- Inserm U955 IMRB, Translational Neuropsychiatry Laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de médecine de précision en psychiatrie (FHU ADAPT), Paris Est Créteil University and Fondation Fondamental, 94010 Créteil, France
| | - Ryad Tamouza
- Inserm U955 IMRB, Translational Neuropsychiatry Laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de médecine de précision en psychiatrie (FHU ADAPT), Paris Est Créteil University and Fondation Fondamental, 94010 Créteil, France
| | - Maher Kharrat
- Laboratory of Human Genetics (LR99ES10), Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
43
|
Baum ML, Wilton DK, Fox RG, Carey A, Hsu YHH, Hu R, Jäntti HJ, Fahey JB, Muthukumar AK, Salla N, Crotty W, Scott-Hewitt N, Bien E, Sabatini DA, Lanser TB, Frouin A, Gergits F, Håvik B, Gialeli C, Nacu E, Lage K, Blom AM, Eggan K, McCarroll SA, Johnson MB, Stevens B. CSMD1 regulates brain complement activity and circuit development. Brain Behav Immun 2024; 119:317-332. [PMID: 38552925 DOI: 10.1016/j.bbi.2024.03.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Complement proteins facilitate synaptic elimination during neurodevelopmental pruning, but neural complement regulation is not well understood. CUB and Sushi Multiple Domains 1 (CSMD1) can regulate complement activity in vitro, is expressed in the brain, and is associated with increased schizophrenia risk. Beyond this, little is known about CSMD1 including whether it regulates complement activity in the brain or otherwise plays a role in neurodevelopment. We used biochemical, immunohistochemical, and proteomic techniques to examine the regional, cellular, and subcellular distribution as well as protein interactions of CSMD1 in the brain. To evaluate whether CSMD1 is involved in complement-mediated synapse elimination, we examined Csmd1-knockout mice and CSMD1-knockout human stem cell-derived neurons. We interrogated synapse and circuit development of the mouse visual thalamus, a process that involves complement pathway activity. We also quantified complement deposition on synapses in mouse visual thalamus and on cultured human neurons. Finally, we assessed uptake of synaptosomes by cultured microglia. We found that CSMD1 is present at synapses and interacts with complement proteins in the brain. Mice lacking Csmd1 displayed increased levels of complement component C3, an increased colocalization of C3 with presynaptic terminals, fewer retinogeniculate synapses, and aberrant segregation of eye-specific retinal inputs to the visual thalamus during the critical period of complement-dependent refinement of this circuit. Loss of CSMD1 in vivo enhanced synaptosome engulfment by microglia in vitro, and this effect was dependent on activity of the microglial complement receptor, CR3. Finally, human stem cell-derived neurons lacking CSMD1 were more vulnerable to complement deposition. These data suggest that CSMD1 can function as a regulator of complement-mediated synapse elimination in the brain during development.
Collapse
Affiliation(s)
- Matthew L Baum
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; MD-PhD Program of Harvard & MIT, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel K Wilton
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rachel G Fox
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alanna Carey
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Han H Hsu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ruilong Hu
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Henna J Jäntti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jaclyn B Fahey
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Allie K Muthukumar
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nikkita Salla
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - William Crotty
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Nicole Scott-Hewitt
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth Bien
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - David A Sabatini
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Toby B Lanser
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Arnaud Frouin
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Frederick Gergits
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Chrysostomi Gialeli
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, S-214 28 Malmö, Sweden; Cardiovascular Research - Translational Studies Research Group, Department of Clinical Sciences, Lund University, S-214 28 Malmö, Sweden
| | - Eugene Nacu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kasper Lage
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, S-214 28 Malmö, Sweden
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Steven A McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Matthew B Johnson
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, USA.
| |
Collapse
|
44
|
Martinez B, Peplow PV. MicroRNAs as potential biomarkers for diagnosis of schizophrenia and influence of antipsychotic treatment. Neural Regen Res 2024; 19:1523-1531. [PMID: 38051895 PMCID: PMC10883514 DOI: 10.4103/1673-5374.387966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/26/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Characterized by positive symptoms (such as changes in behavior or thoughts, including delusions and hallucinations), negative symptoms (such as apathy, anhedonia, and social withdrawal), and cognitive impairments, schizophrenia is a chronic, severe, and disabling mental disorder with late adolescence or early adulthood onset. Antipsychotics are the most commonly used drugs to treat schizophrenia, but those currently in use do not fully reverse all three types of symptoms characterizing this condition. Schizophrenia is frequently misdiagnosed, resulting in a delay of or inappropriate treatment. Abnormal expression of microRNAs is connected to brain development and disease and could provide novel biomarkers for the diagnosis and prognosis of schizophrenia. The recent studies reviewed included microRNA profiling in blood- and urine-based materials and nervous tissue materials. From the studies that had validated the preliminary findings, potential candidate biomarkers for schizophrenia in adults could be miR-22-3p, -30e-5p, -92a-3p, -148b-5p, -181a-3p, -181a-5p, -181b-5p, -199b-5p, -137 in whole blood, and miR-130b, -193a-3p in blood plasma. Antipsychotic treatment of schizophrenia patients was found to modulate the expression of certain microRNAs including miR-130b, -193a-3p, -132, -195, -30e, -432 in blood plasma. Further studies are warranted with adolescents and young adults having schizophrenia and consideration should be given to using animal models of the disorder to investigate the effect of suppressing or overexpressing specific microRNAs.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Pharmacology, University of Nevada-Reno, Reno, NV, USA
- Department of Medicine, University of Nevada-Reno, Reno, NV, USA
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
45
|
Cheng Y, Chen X, Zhang XQ, Ju PJ, Wang WD, Fang Y, Lin GN, Cui DH. Interaction between RNF4 and SART3 is associated with the risk of schizophrenia. Heliyon 2024; 10:e32743. [PMID: 38975171 PMCID: PMC11226853 DOI: 10.1016/j.heliyon.2024.e32743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
The pathogenesis of schizophrenia (SCZ) is heavily influenced by genetic factors. Ring finger protein 4 (RNF4) and squamous cell carcinoma antigen recognized by T cells 3 (SART3) are thought to be involved in nervous system growth and development via oxidative stress pathways. Moreover, they have previously been linked to SCZ. Yet the role of RNF4 and SART3 in SCZ remains unclear. Here, we investigated how these two genes are involved in SCZ by studying their variants observed in patients. We first observed significantly elevated mRNA levels of RNF4 and SART3 in the peripheral blood in both first-episode (n = 30) and chronic (n = 30) SCZ patients compared to controls (n = 60). Next, we targeted-sequenced three single nucleotide polymorphisms (SNPs) in SART3 and six SNPs in RNF4 for association with SCZ using the genomic DNA extracted from peripheral blood leukocytes from SCZ participants (n = 392) and controls (n = 572). We observed a combination of SNPs that included rs1203860, rs2282765 (both in RNF4), and rs2287550 (in SART3) was associated with increased risk of SCZ, suggesting common pathogenic mechanisms between these two genes. We then conducted experiments in HEK293T cells to better understand the interaction between RNF4 and SART3. We observed that SART3 lowered the expression of RNF4 through ubiquitination and downregulated the expression of nuclear factor E2-related factor 2 (NRF2), a downstream factor of RNF4, implicating the existence of a possible shared regulatory mechanism for RNF4 and SART3. In conclusion, our study provides evidence that the interaction between RNF4 and SART3 contributes to the risk of SCZ. The findings shed light on the underlying molecular mechanisms of SCZ and may lead to the development of new therapies and interventions for this disorder.
Collapse
Affiliation(s)
- Ying Cheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Xi Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Xiao Qing Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Pei Jun Ju
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Di Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Division of Imaging, Computational and Systems Biomedicine, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Fang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Guan Ning Lin
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Division of Imaging, Computational and Systems Biomedicine, School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Dong Hong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
- Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
46
|
Bodai L, Borosta R, Ferencz Á, Kovács M, Zsindely N. The Role of miR-137 in Neurodegenerative Disorders. Int J Mol Sci 2024; 25:7229. [PMID: 39000336 PMCID: PMC11241563 DOI: 10.3390/ijms25137229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases affect an increasing part of the population of modern societies, burdening healthcare systems and causing immense suffering at the personal level. The pathogenesis of several of these disorders involves dysregulation of gene expression, which depends on several molecular processes ranging from transcription to protein stability. microRNAs (miRNAs) are short non-coding RNA molecules that modulate gene expression by suppressing the translation of partially complementary mRNAs. miR-137 is a conserved, neuronally enriched miRNA that is implicated in neurodegeneration. Here, we review the current body of knowledge about the role that miR-137 plays in five prominent neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The presented data indicate that, rather than having a general neuroprotective role, miR-137 modulates the pathology of distinct disorders differently.
Collapse
Affiliation(s)
- László Bodai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Roberta Borosta
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Ágnes Ferencz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Mercédesz Kovács
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Nóra Zsindely
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
- Department of Genetics, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| |
Collapse
|
47
|
Lawn T, Giacomel A, Martins D, Veronese M, Howard M, Turkheimer FE, Dipasquale O. Normative modelling of molecular-based functional circuits captures clinical heterogeneity transdiagnostically in psychiatric patients. Commun Biol 2024; 7:689. [PMID: 38839931 PMCID: PMC11153627 DOI: 10.1038/s42003-024-06391-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/27/2024] [Indexed: 06/07/2024] Open
Abstract
Advanced methods such as REACT have allowed the integration of fMRI with the brain's receptor landscape, providing novel insights transcending the multiscale organisation of the brain. Similarly, normative modelling has allowed translational neuroscience to move beyond group-average differences and characterise deviations from health at an individual level. Here, we bring these methods together for the first time. We used REACT to create functional networks enriched with the main modulatory, inhibitory, and excitatory neurotransmitter systems and generated normative models of these networks to capture functional connectivity deviations in patients with schizophrenia, bipolar disorder (BPD), and ADHD. Substantial overlap was seen in symptomatology and deviations from normality across groups, but these could be mapped into a common space linking constellations of symptoms through to underlying neurobiology transdiagnostically. This work provides impetus for developing novel biomarkers that characterise molecular- and systems-level dysfunction at the individual level, facilitating the transition towards mechanistically targeted treatments.
Collapse
Affiliation(s)
- Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Alessio Giacomel
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Matthew Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Department of Research & Development Advanced Applications, Olea Medical, La Ciotat, France.
| |
Collapse
|
48
|
Yan Y, Dai T, Guo M, Zhao X, Chen C, Zhou Y, Qin M, Xu L, Zhao J. A review of non-classical MAPK family member, MAPK4: A pivotal player in cancer development and therapeutic intervention. Int J Biol Macromol 2024; 271:132686. [PMID: 38801852 DOI: 10.1016/j.ijbiomac.2024.132686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Mitogen-Activated Protein Kinases (MAPKs) are serine/threonine protein kinases that play a crucial role in transmitting extracellular signals to the intracellular environment, influencing a wide range of cellular processes including proliferation, differentiation, apoptosis, metabolic activities, immune function and stress response. MAPK4, a non-classical MAPK, is frequently overexpressed in various malignancies, including prostate, breast, cervix, thyroid, and gliomas. It orchestrates cell proliferation, migration, and apoptosis via the AKT/mTOR and/or PDK1 signaling pathways, thus facilitating tumor cell growth. Furthermore, MAPK4 expression is closely associated with the effectiveness of specific inhibitors like PI3K and PARP1, and also correlate with the survival rates of cancer patients. Increasing evidence highlights MAPK4's involvement in the tumor microenvironment, modulating immune response and inflammation-related diseases. This review comprehensively explores the structure, function, and oncogenic role of MAPK4, providing a deeper understanding of its activation and mechanisms of action in tumorigenesis, which might be helpful for the development of innovative therapeutic strategies for cancer management.
Collapse
Affiliation(s)
- Yaping Yan
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Tengkun Dai
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Mengmeng Guo
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Xu Zhao
- Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China; School of Medicine, Guizhou University, Guiyang 550025, Guizhou, China
| | - Chao Chen
- Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China; School of Medicine, Guizhou University, Guiyang 550025, Guizhou, China
| | - Ya Zhou
- Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China; Department of Medical physics, Zunyi Medical University, Guizhou 563000, China
| | - Ming Qin
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China
| | - Lin Xu
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China.
| | - Juanjuan Zhao
- Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Key Laboratory of Gene Detection and Treatment of Guizhou province, Zunyi 563000, China.
| |
Collapse
|
49
|
Zhu ZH, Yin XY, Cai Y, Jia NN, Wang PJ, Qi Q, Hou WL, Man LJ, Hui L. Association between the HHEX polymorphism and delayed memory in first-episode schizophrenic patients. Schizophr Res Cogn 2024; 36:100304. [PMID: 38444400 PMCID: PMC10912683 DOI: 10.1016/j.scog.2024.100304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/23/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
The hematopoietically-expressed homeobox gene (HHEX) played a critical role in regulating the immune system that the abnormality of which was involved in the psychopathology and cognitive deficits of psychiatric disorders. The aim of this study was to investigate the effect of HHEX rs1111875 polymorphism on the susceptibility and cognitive deficits of first-episode schizophrenic patients (FSP). We assessed cognitive function in 239 first-episode patients meeting DSM-IV for schizophrenia, and 368 healthy controls using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). The HHEX rs1111875 polymorphism was genotyped. Our results showed that the allelic and genotypic frequencies of HHEX rs1111875 polymorphism didn't differ between FSP and healthy controls (both p > 0.05) after adjusting for sex and age. Cognitive test scores in FSP were significantly lower than those in healthy controls on all scales (all p < 0.001) except for the visuospatial/constructional score (p > 0.05) after adjusting for covariates. There was a significant genotype (p < 0.05) rather than genotype × diagnosis (p > 0.05) effect on the delayed memory score after adjusting for covariates. The HHEX rs1111875 polymorphism was significantly associated with the delayed memory score in FSP (p < 0.05), but not in healthy controls (p > 0.05) after adjusting for covariates. Our findings supported that the HHEX rs1111875 polymorphism did not contribute to the susceptibility to FSP. However, this polymorphism might influence the delayed memory in FSP. Moreover, FSP had poorer cognitive function than healthy controls except for the visuospatial/constructional domain.
Collapse
Affiliation(s)
| | | | | | - Ning Ning Jia
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Pei Jie Wang
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Qi Qi
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Wen Long Hou
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Li Juan Man
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou 215137, Jiangsu, PR China
| |
Collapse
|
50
|
Werren EA, Peirent ER, Jantti H, Guxholli A, Srivastava KR, Orenstein N, Narayanan V, Wiszniewski W, Dawidziuk M, Gawlinski P, Umair M, Khan A, Khan SN, Geneviève D, Lehalle D, van Gassen KLI, Giltay JC, Oegema R, van Jaarsveld RH, Rafiullah R, Rappold GA, Rabin R, Pappas JG, Wheeler MM, Bamshad MJ, Tsan YC, Johnson MB, Keegan CE, Srivastava A, Bielas SL. Biallelic variants in CSMD1 are implicated in a neurodevelopmental disorder with intellectual disability and variable cortical malformations. Cell Death Dis 2024; 15:379. [PMID: 38816421 PMCID: PMC11140003 DOI: 10.1038/s41419-024-06768-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 05/03/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
CSMD1 (Cub and Sushi Multiple Domains 1) is a well-recognized regulator of the complement cascade, an important component of the innate immune response. CSMD1 is highly expressed in the central nervous system (CNS) where emergent functions of the complement pathway modulate neural development and synaptic activity. While a genetic risk factor for neuropsychiatric disorders, the role of CSMD1 in neurodevelopmental disorders is unclear. Through international variant sharing, we identified inherited biallelic CSMD1 variants in eight individuals from six families of diverse ancestry who present with global developmental delay, intellectual disability, microcephaly, and polymicrogyria. We modeled CSMD1 loss-of-function (LOF) pathogenesis in early-stage forebrain organoids differentiated from CSMD1 knockout human embryonic stem cells (hESCs). We show that CSMD1 is necessary for neuroepithelial cytoarchitecture and synchronous differentiation. In summary, we identified a critical role for CSMD1 in brain development and biallelic CSMD1 variants as the molecular basis of a previously undefined neurodevelopmental disorder.
Collapse
Affiliation(s)
- Elizabeth A Werren
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Advanced Precision Medicine Laboratory, The Jackson Laboratory for Genomic Medicine, Farmington, CTt, 06032, USA
| | - Emily R Peirent
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Henna Jantti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Alba Guxholli
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Kinshuk Raj Srivastava
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Naama Orenstein
- Schneider Children's Medical Center of Israel, Petah Tikva, 4920235, Israel
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Wojciech Wiszniewski
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Mateusz Dawidziuk
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, 01-211, Poland
| | - Pawel Gawlinski
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, 01-211, Poland
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, 11481, Saudi Arabia
- Department of Life Sciences, School of Science, University of Management and Technology, Lahore, Punjab, 54770, Pakistan
| | - Amjad Khan
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Zoology, University of Lakki Marwat, Lakki Marwat, Khyber Pakhtunkhwa, 28420, Pakistan
| | - Shahid Niaz Khan
- Department of Zoology, Kohat University of Science and Technology, Kohat, Pakistan
| | - David Geneviève
- Montpellier University, Inserm Unit U1183, Reference Center for Rare Diseases and Developmental Anomalies, CHU, 34000, Montpellier, France
| | - Daphné Lehalle
- Sorbonne University, Department of Medical Genetics, Hospital Armand Trousseau, 75012, Paris, France
| | - K L I van Gassen
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, 3584 EA, The Netherlands
| | - Jacques C Giltay
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, 3584 EA, The Netherlands
| | - Renske Oegema
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, 3584 EA, The Netherlands
| | - Richard H van Jaarsveld
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, 3584 EA, The Netherlands
| | - Rafiullah Rafiullah
- Department of Biotechnology, Faculty of Life Sciences, BUITEMS, Quetta, 87300, Pakistan
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Ruprecht-Karls-University, Heidelberg, 69120, Germany
| | - Rachel Rabin
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - John G Pappas
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Marsha M Wheeler
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
- Brotman Baty Institute, Washington, 98195, USA
| | - Yao-Chang Tsan
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthew B Johnson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Catherine E Keegan
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Anshika Srivastava
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India.
| | - Stephanie L Bielas
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|