1
|
Sharma D, Bharadaj SK, Bharadaj S, Chakraborty S. MicroRNA-regulated suppression of some overexpressed genes in schizophrenia and their evolutionary significance. Schizophr Res 2025; 276:143-156. [PMID: 39892248 DOI: 10.1016/j.schres.2025.01.022] [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: 07/12/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Impaired formation of the brain or nervous system is the root cause of neurodevelopmental disorders which appear in pregnancy or soon after birth. One such neurodevelopmental disorder is Schizophrenia. Among the most serious forms of psychosis, Schizophrenia (SCZ) affects 1 % of the general population. MiRNA targeting sites and codon usage of nine overexpressed genes in Schizophrenia were investigated in this study. The neutrality plot demonstrated the importance of natural selection over mutational pressure in the evolution of these genes. The analysis of COSM revealed that the miRNA target regions of the genes were encoded by non-optimal codons, resulting in low translational efficiency, whereas the GC content revealed that the miRNA-mRNA binding was strong. From this study it was concluded that each overexpressed gene was targeted by several human miRNAs for putative suppression. These identified miRNAs could have therapeutic potential in Schizophrenia therapy.
Collapse
Affiliation(s)
- Deepika Sharma
- Department of Biotechnology, Assam University, Silchar 788011, Assam, India
| | | | - Stella Bharadaj
- Silchar Medical College and Hospital, Silchar 788014, Assam, India
| | - Supriyo Chakraborty
- Department of Biotechnology, Assam University, Silchar 788011, Assam, India.
| |
Collapse
|
2
|
Sportelli L, Eisenberg DP, Passiatore R, D'Ambrosio E, Antonucci LA, Bettina JS, Chen Q, Goldman AL, Gregory MD, Griffiths K, Hyde TM, Kleinman JE, Pardiñas AF, Parihar M, Popolizio T, Rampino A, Shin JH, Veronese M, Ulrich WS, Zink CF, Bertolino A, Howes OD, Berman KF, Weinberger DR, Pergola G. Dopamine signaling enriched striatal gene set predicts striatal dopamine synthesis and physiological activity in vivo. Nat Commun 2024; 15:3342. [PMID: 38688917 PMCID: PMC11061310 DOI: 10.1038/s41467-024-47456-5] [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: 09/04/2023] [Accepted: 03/22/2024] [Indexed: 05/02/2024] Open
Abstract
The polygenic architecture of schizophrenia implicates several molecular pathways involved in synaptic function. However, it is unclear how polygenic risk funnels through these pathways to translate into syndromic illness. Using tensor decomposition, we analyze gene co-expression in the caudate nucleus, hippocampus, and dorsolateral prefrontal cortex of post-mortem brain samples from 358 individuals. We identify a set of genes predominantly expressed in the caudate nucleus and associated with both clinical state and genetic risk for schizophrenia that shows dopaminergic selectivity. A higher polygenic risk score for schizophrenia parsed by this set of genes predicts greater dopamine synthesis in the striatum and greater striatal activation during reward anticipation. These results translate dopamine-linked genetic risk variation into in vivo neurochemical and hemodynamic phenotypes in the striatum that have long been implicated in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Leonardo Sportelli
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Daniel P Eisenberg
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, NIH, DHHS, Bethesda, MD, USA
| | - Roberta Passiatore
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Enrico D'Ambrosio
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Linda A Antonucci
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Jasmine S Bettina
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, NIH, DHHS, Bethesda, MD, USA
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Aaron L Goldman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Michael D Gregory
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, NIH, DHHS, Bethesda, MD, USA
| | - Kira Griffiths
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
- Holmusk Technologies, New York, NY, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Madhur Parihar
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Teresa Popolizio
- Radiology Department, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Antonio Rampino
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
- Azienda Ospedaliero Universitaria Consorziale Policlinico, Bari, Italy
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Mattia Veronese
- Department of Information Engineering, University of Padua, Padua, Italy
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - William S Ulrich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Caroline F Zink
- Baltimore Research and Education Foundation, Baltimore, MD, USA
| | - Alessandro Bertolino
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
- Azienda Ospedaliero Universitaria Consorziale Policlinico, Bari, Italy
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Karen F Berman
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, NIH, DHHS, Bethesda, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Giulio Pergola
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA.
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Polakkattil BK, Vellichirammal NN, Nair IV, Nair CM, Banerjee M. Methylome-wide and meQTL analysis helps to distinguish treatment response from non-response and pathogenesis markers in schizophrenia. Front Psychiatry 2024; 15:1297760. [PMID: 38516266 PMCID: PMC10954811 DOI: 10.3389/fpsyt.2024.1297760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/06/2024] [Indexed: 03/23/2024] Open
Abstract
Schizophrenia is a complex condition with entwined genetic and epigenetic risk factors, posing a challenge to disentangle the intermixed pathological and therapeutic epigenetic signatures. To resolve this, we performed 850K methylome-wide and 700K genome-wide studies on the same set of schizophrenia patients by stratifying them into responders, non-responders, and drug-naïve patients. The key genes that signified the response were followed up using real-time gene expression studies to understand the effect of antipsychotics at the gene transcription level. The study primarily implicates hypermethylation in therapeutic response and hypomethylation in the drug-non-responsive state. Several differentially methylated sites and regions colocalized with the schizophrenia genome-wide association study (GWAS) risk genes and variants, supporting the convoluted gene-environment association. Gene ontology and protein-protein interaction (PPI) network analyses revealed distinct patterns that differentiated the treatment response from drug resistance. The study highlights the strong involvement of several processes related to nervous system development, cell adhesion, and signaling in the antipsychotic response. The ability of antipsychotic medications to alter the pathology by modulating gene expression or methylation patterns is evident from the general increase in the gene expression of response markers and histone modifiers and the decrease in class II human leukocyte antigen (HLA) genes following treatment with varying concentrations of medications like clozapine, olanzapine, risperidone, and haloperidol. The study indicates a directional overlap of methylation markers between pathogenesis and therapeutic response, thereby suggesting a careful distinction of methylation markers of pathogenesis from treatment response. In addition, there is a need to understand the trade-off between genetic and epigenetic observations. It is suggested that methylomic changes brought about by drugs need careful evaluation for their positive effects on pathogenesis, course of disease progression, symptom severity, side effects, and refractoriness.
Collapse
Affiliation(s)
- Binithamol K. Polakkattil
- Human Molecular Genetics Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
- Research Center, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Neetha N. Vellichirammal
- Human Molecular Genetics Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Indu V. Nair
- Mental Health Centre, Thiruvananthapuram, Kerala, India
| | | | - Moinak Banerjee
- Human Molecular Genetics Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
4
|
Sportelli L, Eisenberg DP, Passiatore R, D'Ambrosio E, Antonucci LA, Chen Q, Czarapata J, Goldman AL, Gregory M, Griffiths K, Hyde TM, Kleinman JE, Pardiñas AF, Parihar M, Popolizio T, Rampino A, Shin JH, Veronese M, Ulrich WS, Zink CF, Bertolino A, Howes OD, Berman KF, Weinberger DR, Pergola G. Dopamine and schizophrenia from bench to bedside: Discovery of a striatal co-expression risk gene set that predicts in vivo measures of striatal function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558594. [PMID: 37786720 PMCID: PMC10541621 DOI: 10.1101/2023.09.20.558594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Schizophrenia (SCZ) is characterized by a polygenic risk architecture implicating diverse molecular pathways important for synaptic function. However, how polygenic risk funnels through these pathways to translate into syndromic illness is unanswered. To evaluate biologically meaningful pathways of risk, we used tensor decomposition to characterize gene co-expression in post-mortem brain (of neurotypicals: N=154; patients with SCZ: N=84; and GTEX samples N=120) from caudate nucleus (CN), hippocampus (HP), and dorsolateral prefrontal cortex (DLPFC). We identified a CN-predominant gene set showing dopaminergic selectivity that was enriched for genes associated with clinical state and for genes associated with SCZ risk. Parsing polygenic risk score for SCZ based on this specific gene set (parsed-PRS), we found that greater pathway-specific SCZ risk predicted greater in vivo striatal dopamine synthesis capacity measured by [ 18 F]-FDOPA PET in three independent cohorts of neurotypicals and patients (total N=235) and greater fMRI striatal activation during reward anticipation in two additional independent neurotypical cohorts (total N=141). These results reveal a 'bench to bedside' translation of dopamine-linked genetic risk variation in driving in vivo striatal neurochemical and hemodynamic phenotypes that have long been implicated in the pathophysiology of SCZ.
Collapse
|
5
|
Pergola G, Parihar M, Sportelli L, Bharadwaj R, Borcuk C, Radulescu E, Bellantuono L, Blasi G, Chen Q, Kleinman JE, Wang Y, Sripathy SR, Maher BJ, Monaco A, Rossi F, Shin JH, Hyde TM, Bertolino A, Weinberger DR. Consensus molecular environment of schizophrenia risk genes in coexpression networks shifting across age and brain regions. SCIENCE ADVANCES 2023; 9:eade2812. [PMID: 37058565 PMCID: PMC10104472 DOI: 10.1126/sciadv.ade2812] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 03/10/2023] [Indexed: 06/19/2023]
Abstract
Schizophrenia is a neurodevelopmental brain disorder whose genetic risk is associated with shifting clinical phenomena across the life span. We investigated the convergence of putative schizophrenia risk genes in brain coexpression networks in postmortem human prefrontal cortex (DLPFC), hippocampus, caudate nucleus, and dentate gyrus granule cells, parsed by specific age periods (total N = 833). The results support an early prefrontal involvement in the biology underlying schizophrenia and reveal a dynamic interplay of regions in which age parsing explains more variance in schizophrenia risk compared to lumping all age periods together. Across multiple data sources and publications, we identify 28 genes that are the most consistently found partners in modules enriched for schizophrenia risk genes in DLPFC; twenty-three are previously unidentified associations with schizophrenia. In iPSC-derived neurons, the relationship of these genes with schizophrenia risk genes is maintained. The genetic architecture of schizophrenia is embedded in shifting coexpression patterns across brain regions and time, potentially underwriting its shifting clinical presentation.
Collapse
Affiliation(s)
- Giulio Pergola
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Madhur Parihar
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Leonardo Sportelli
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Rahul Bharadwaj
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Christopher Borcuk
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Eugenia Radulescu
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Loredana Bellantuono
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
- Istituto Nazionale di Fisica Nucleare, Bari, Italy
| | - Giuseppe Blasi
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
- Azienda Ospedaliero Universitaria Consorziale Policlinico, Bari, Italy
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yanhong Wang
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Srinidhi Rao Sripathy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Brady J. Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alfonso Monaco
- Istituto Nazionale di Fisica Nucleare, Bari, Italy
- Dipartimento Interateneo di Fisica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Fabiana Rossi
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alessandro Bertolino
- Group of Psychiatric Neuroscience, Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
- Azienda Ospedaliero Universitaria Consorziale Policlinico, Bari, Italy
| | - Daniel R. Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Panariello F, Fanelli G, Fabbri C, Atti AR, De Ronchi D, Serretti A. Epigenetic Basis of Psychiatric Disorders: A Narrative Review. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:302-315. [PMID: 34433406 DOI: 10.2174/1871527320666210825101915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/02/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Psychiatric disorders are complex, multifactorial illnesses with a demonstrated biological component in their etiopathogenesis. Epigenetic modifications, through the modulation of DNA methylation, histone modifications and RNA interference, tune tissue-specific gene expression patterns and play a relevant role in the etiology of psychiatric illnesses. OBJECTIVE This review aims to discuss the epigenetic mechanisms involved in psychiatric disorders, their modulation by environmental factors and their interactions with genetic variants, in order to provide a comprehensive picture of their mutual crosstalk. METHODS In accordance with the PRISMA guidelines, systematic searches of Medline, EMBASE, PsycINFO, Web of Science, Scopus, and the Cochrane Library were conducted. RESULTS Exposure to environmental factors, such as poor socio-economic status, obstetric complications, migration, and early life stressors, may lead to stable changes in gene expression and neural circuit function, playing a role in the risk of psychiatric diseases. The most replicated genes involved by studies using different techniques are discussed. Increasing evidence indicates that these sustained abnormalities are maintained by epigenetic modifications in specific brain regions and they interact with genetic variants in determining the risk of psychiatric disorders. CONCLUSION An increasing amount of evidence suggests that epigenetics plays a pivotal role in the etiopathogenesis of psychiatric disorders. New therapeutic approaches may work by reversing detrimental epigenetic changes that occurred during the lifespan.
Collapse
Affiliation(s)
- Fabio Panariello
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Fanelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Rita Atti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Diana De Ronchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
7
|
Wagh VV, Vyas P, Agrawal S, Pachpor TA, Paralikar V, Khare SP. Peripheral Blood-Based Gene Expression Studies in Schizophrenia: A Systematic Review. Front Genet 2021; 12:736483. [PMID: 34721526 PMCID: PMC8548640 DOI: 10.3389/fgene.2021.736483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022] Open
Abstract
Schizophrenia is a disorder that is characterized by delusions, hallucinations, disorganized speech or behavior, and socio-occupational impairment. The duration of observation and variability in symptoms can make the accurate diagnosis difficult. Identification of biomarkers for schizophrenia (SCZ) can help in early diagnosis, ascertaining the diagnosis, and development of effective treatment strategies. Here we review peripheral blood-based gene expression studies for identification of gene expression biomarkers for SCZ. A literature search was carried out in PubMed and Web of Science databases for blood-based gene expression studies in SCZ. A list of differentially expressed genes (DEGs) was compiled and analyzed for overlap with genetic markers, differences based on drug status of the participants, functional enrichment, and for effect of antipsychotics. This literature survey identified 61 gene expression studies. Seventeen out of these studies were based on expression microarrays. A comparative analysis of the DEGs (n = 227) from microarray studies revealed differences between drug-naive and drug-treated SCZ participants. We found that of the 227 DEGs, 11 genes (ACOT7, AGO2, DISC1, LDB1, RUNX3, SIGIRR, SLC18A1, NRG1, CHRNB2, PRKAB2, and ZNF74) also showed genetic and epigenetic changes associated with SCZ. Functional enrichment analysis of the DEGs revealed dysregulation of proline and 4-hydroxyproline metabolism. Also, arginine and proline metabolism was the most functionally enriched pathway for SCZ in our analysis. Follow-up studies identified effect of antipsychotic treatment on peripheral blood gene expression. Of the 27 genes compiled from the follow-up studies AKT1, DISC1, HP, and EIF2D had no effect on their expression status as a result of antipsychotic treatment. Despite the differences in the nature of the study, ethnicity of the population, and the gene expression analysis method used, we identified several coherent observations. An overlap, though limited, of genetic, epigenetic and gene expression changes supports interplay of genetic and environmental factors in SCZ. The studies validate the use of blood as a surrogate tissue for biomarker analysis. We conclude that well-designed cohort studies across diverse populations, use of high-throughput sequencing technology, and use of artificial intelligence (AI) based computational analysis will significantly improve our understanding and diagnostic capabilities for this complex disorder.
Collapse
Affiliation(s)
- Vipul Vilas Wagh
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Parin Vyas
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Suchita Agrawal
- The Psychiatry Unit, KEM Hospital and KEM Hospital Research Centre, Pune, India
| | | | - Vasudeo Paralikar
- The Psychiatry Unit, KEM Hospital and KEM Hospital Research Centre, Pune, India
| | - Satyajeet P Khare
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
8
|
Shen L, Lv X, Huang H, Li M, Huai C, Wu X, Wu H, Ma J, Chen L, Wang T, Tan J, Sun Y, Li L, Shi Y, Yang C, Cai L, Lu Y, Zhang Y, Weng S, Tai S, Zhang N, He L, Wan C, Qin S. Genome-wide analysis of DNA methylation in 106 schizophrenia family trios in Han Chinese. EBioMedicine 2021; 72:103609. [PMID: 34628353 PMCID: PMC8511801 DOI: 10.1016/j.ebiom.2021.103609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/05/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022] Open
Abstract
Background Schizophrenia (SCZ) is a severe psychiatric disorder that affects approximately 0.75% of the global population. Both genetic and environmental factors contribute to development of SCZ. SCZ tends to run in family while both genetic and environmental factor contribute to its etiology. Much evidence suggested that alterations in DNA methylations occurred in SCZ patients. Methods To investigate potential inheritable pattern of DNA methylation in SCZ family, we performed a genome-wide analysis of DNA methylation of peripheral blood samples from 106 Chinese SCZ family trios. Genome-wide DNA methylations were quantified by Agilent 1 × 244 k Human Methylation Microarray. Findings In this study, we proposed a loci inheritance frequency model that allows characterization of differential methylated regions as SCZ biomarkers. Based on this model, 112 hypermethylated and 125 hypomethylated regions were identified. Additionally, 121 hypermethylated and 139 hypomethylated genes were annotated. The results of functional enrichment analysis indicated that multiple differentially methylated genes (DMGs) involved in Notch/HH/Wnt signaling, MAPK signaling, GPCR signaling, immune response signaling. Notably, a number of hypomethylated genes were significantly enriched in cerebral cortex and functionally enriched in nervous system development. Interpretation Our findings not only validated previously discovered risk genes of SCZ but also identified novel candidate DMGs in SCZ. These results may further the understanding of altered DNA methylations in SCZ.
Collapse
Affiliation(s)
- Lu Shen
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoying Lv
- DCH Technologies Inc, Cambridge, MA 02142, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences, Shanghai 200031, PR China; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mo Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Cong Huai
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Xi Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Hao Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Jingsong Ma
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Luan Chen
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Ting Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Jie Tan
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Yidan Sun
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lixing Li
- Department of General Surgery, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Shi
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Chao Yang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lei Cai
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Yana Lu
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi 214151, China
| | - Yan Zhang
- The Second People's Hospital of Lishui, Lishui 323020, China
| | - Saizheng Weng
- Fuzhou Neuro-psychiatric hospital, Fujian Medical University, Fuzhou 350026, China
| | - Shaobin Tai
- The Second People's Hospital of Huangshan, Huangshan 245021, China
| | - Na Zhang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lin He
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China; Department of General Surgery, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| | - Chunling Wan
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| | - Shengying Qin
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| |
Collapse
|
9
|
Nie FY, Zhang MR, Shang SS, Zhang QX, Zhang R, Chen P, Ma J. Methylome-wide association study of first-episode schizophrenia reveals a hypermethylated CpG site in the promoter region of the TNIK susceptibility gene. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110081. [PMID: 32853717 DOI: 10.1016/j.pnpbp.2020.110081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022]
Abstract
Accumulating evidence suggests that epigenetics plays an important role in the etiology of schizophrenia. Here, we performed a methylome-wide association study (MWAS) of first-onset schizophrenia patients and controls from the Han Chinese population using microarray technology. The DNA methylation profiles revealed 4494 differentially methylated CpG sites. Gene ontology (GO) analysis showed that the functions of differentially methylated genes were primarily involved in enzymatic activity, cytoskeleton organization and cell adhesion, and the TNIK (encoding TRAF2- and NCK-interacting kinase) gene was enriched in most of these terms. By combining the MWAS results with those of previous genome-wide association studies (GWASs), we identified 72 candidate genes located in 49 human genome loci. Among the overlapping genes, the most significantly methylated CpG sites were in the transcriptional start site (TSS) 200 region (cg21413905, Punadjusted = 3.20 × 10-5) of TNIK. TNIK was listed in the top 50 differentially methylated loci. The results of pyrosequencing and TNIK mRNA expression were consistent with those of the microarray study. Bioinformatics analyses, dual-luciferase reporter assays and chromatin immunoprecipitation (ChIP) studies showed that TNIK interacted with genes associated with schizophrenia and NRF1 was identified as a novel transcription factor (TF) that binds to TNIK in its TSS200 region. Thus, the regulatory function of NRF1 may be influenced by the status of the methylated CpG site in this region. In summary, our study provides new insights into the epigenetic mechanisms that regulate schizophrenia. Studies of the functions of TNIK methylation should be performed in vitro and in vivo to provide a better understanding of the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Fa-Yi Nie
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Medical Research Center, Xi'an No.3 Hospital, Xi'an, Shaanxi 710018, China
| | - Miao-Ran Zhang
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Shan-Shan Shang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Qiao-Xia Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Rui Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Peng Chen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| | - Jie Ma
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Medical Research Center, Xi'an No.3 Hospital, Xi'an, Shaanxi 710018, China.
| |
Collapse
|
10
|
Hannon E, Dempster EL, Mansell G, Burrage J, Bass N, Bohlken MM, Corvin A, Curtis CJ, Dempster D, Di Forti M, Dinan TG, Donohoe G, Gaughran F, Gill M, Gillespie A, Gunasinghe C, Hulshoff HE, Hultman CM, Johansson V, Kahn RS, Kaprio J, Kenis G, Kowalec K, MacCabe J, McDonald C, McQuillin A, Morris DW, Murphy KC, Mustard CJ, Nenadic I, O'Donovan MC, Quattrone D, Richards AL, Rutten BPF, St Clair D, Therman S, Toulopoulou T, Van Os J, Waddington JL, Sullivan P, Vassos E, Breen G, Collier DA, Murray RM, Schalkwyk LS, Mill J. DNA methylation meta-analysis reveals cellular alterations in psychosis and markers of treatment-resistant schizophrenia. eLife 2021; 10:e58430. [PMID: 33646943 PMCID: PMC8009672 DOI: 10.7554/elife.58430] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 02/23/2021] [Indexed: 12/30/2022] Open
Abstract
We performed a systematic analysis of blood DNA methylation profiles from 4483 participants from seven independent cohorts identifying differentially methylated positions (DMPs) associated with psychosis, schizophrenia, and treatment-resistant schizophrenia. Psychosis cases were characterized by significant differences in measures of blood cell proportions and elevated smoking exposure derived from the DNA methylation data, with the largest differences seen in treatment-resistant schizophrenia patients. We implemented a stringent pipeline to meta-analyze epigenome-wide association study (EWAS) results across datasets, identifying 95 DMPs associated with psychosis and 1048 DMPs associated with schizophrenia, with evidence of colocalization to regions nominated by genetic association studies of disease. Many schizophrenia-associated DNA methylation differences were only present in patients with treatment-resistant schizophrenia, potentially reflecting exposure to the atypical antipsychotic clozapine. Our results highlight how DNA methylation data can be leveraged to identify physiological (e.g., differential cell counts) and environmental (e.g., smoking) factors associated with psychosis and molecular biomarkers of treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Eilis Hannon
- University of Exeter Medical School, University of Exeter, Barrack RoadExeterUnited Kingdom
| | - Emma L Dempster
- University of Exeter Medical School, University of Exeter, Barrack RoadExeterUnited Kingdom
| | - Georgina Mansell
- University of Exeter Medical School, University of Exeter, Barrack RoadExeterUnited Kingdom
| | - Joe Burrage
- University of Exeter Medical School, University of Exeter, Barrack RoadExeterUnited Kingdom
| | - Nick Bass
- Division of Psychiatry, University College LondonLondonUnited Kingdom
| | - Marc M Bohlken
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, HeidelberglaanUtrechtNetherlands
| | - Aiden Corvin
- Department of Psychiatry and Neuropsychiatric Genetics Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, St. James HospitalDublinIreland
| | - Charles J Curtis
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- NIHR BioResource Centre Maudsley, South London and Maudsley NHS Foundation Trust (SLaM), King’s College LondonLondonUnited Kingdom
| | - David Dempster
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- South London and Maudsley NHS Mental Health Foundation TrustLondonUnited Kingdom
- National Institute for Health Research (NIHR), Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College LondonLondonUnited Kingdom
| | | | - Gary Donohoe
- Centre for Neuroimaging and Cognitive Genomics (NICOG), School of Psychology and Discipline of Biochemistry, National University of Ireland GalwayGalwayIreland
| | - Fiona Gaughran
- Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- National Psychosis Service, South London and Maudsley NHS Foundation TrustLondonUnited Kingdom
| | - Michael Gill
- Department of Psychiatry and Neuropsychiatric Genetics Research Group, Trinity Translational Medicine Institute, Trinity College DublinDublinIreland
| | - Amy Gillespie
- Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- Department of Psychiatry, Medical Sciences Division, University of OxfordOxfordUnited Kingdom
| | - Cerisse Gunasinghe
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
| | - Hilleke E Hulshoff
- Department of Psychiatry, University Medical Center UtrechtUtrechtNetherlands
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska InstitutetStockholmSweden
| | - Viktoria Johansson
- Department of Medical Epidemiology and Biostatistics Sweden, Karolinska InstitutetStockholmSweden
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm Health Care ServicesStockholmSweden
| | - René S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrechtNetherlands
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jaakko Kaprio
- Institute for Molecular Medicine FIMM, University of HelsinkiHelsinkiFinland
- Department of Public Health, University of HelsinkiHelsinkiFinland
| | - Gunter Kenis
- Faculty of Health, Medicine and Life Sciences, Maastricht UniversityMaastrichtNetherlands
| | - Kaarina Kowalec
- Department of Medical Epidemiology and Biostatistics, Karolinska InstitutetStockholmSweden
- College of Pharmacy, University of ManitobaWinnipegCanada
| | - James MacCabe
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
| | - Colm McDonald
- Centre for Neuroimaging and Cognitive Genomics (NICOG), School of Medicine, National University of Ireland GalwayGalwayIreland
| | - Andrew McQuillin
- Division of Psychiatry, University College LondonLondonUnited Kingdom
- Division of Psychiatry, University College LondonLondonUnited Kingdom
| | - Derek W Morris
- Centre for Neuroimaging and Cognitive Genomics (NICOG), School of Psychology and Discipline of Biochemistry, National University of Ireland GalwayGalwayIreland
| | - Kieran C Murphy
- Department of Psychiatry, Royal College of Surgeons in IrelandDublinIreland
| | - Colette J Mustard
- Division of Biomedical Sciences, Institute of Health Research and Innovation, University of the Highlands and IslandsInvernessUnited Kingdom
| | - Igor Nenadic
- Department of Psychiatry and Psychotherapy, Jena University HospitalJenaGermany
- Department of Psychiatry and Psychotherapy, Philipps University Marburg/ Marburg University Hospital UKGMMarburgGermany
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Diego Quattrone
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- South London and Maudsley NHS Mental Health Foundation TrustLondonUnited Kingdom
| | - Alexander L Richards
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Bart PF Rutten
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht UniversityMaastrichtNetherlands
| | - David St Clair
- The Institute of Medical Sciences, Univeristy of AberdeenAberdeenUnited Kingdom
| | - Sebastian Therman
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and WelfareHelsinkiFinland
| | - Timothea Toulopoulou
- Department of Psychology and National Magnetic Resonance Research Center (UMRAM), Aysel Sabuncu Brain Research Centre (ASBAM), Bilkent UniversityAnkaraTurkey
| | - Jim Van Os
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrechtNetherlands
| | - John L Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in IrelandDublinIreland
| | - Wellcome Trust Case Control Consortium (WTCCC)
- University of Exeter Medical School, University of Exeter, Barrack RoadExeterUnited Kingdom
- Division of Psychiatry, University College LondonLondonUnited Kingdom
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, HeidelberglaanUtrechtNetherlands
- Department of Psychiatry and Neuropsychiatric Genetics Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, St. James HospitalDublinIreland
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- NIHR BioResource Centre Maudsley, South London and Maudsley NHS Foundation Trust (SLaM), King’s College LondonLondonUnited Kingdom
- South London and Maudsley NHS Mental Health Foundation TrustLondonUnited Kingdom
- National Institute for Health Research (NIHR), Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College LondonLondonUnited Kingdom
- APC Microbiome Ireland, University College CorkCorkIreland
- Centre for Neuroimaging and Cognitive Genomics (NICOG), School of Psychology and Discipline of Biochemistry, National University of Ireland GalwayGalwayIreland
- Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- National Psychosis Service, South London and Maudsley NHS Foundation TrustLondonUnited Kingdom
- Department of Psychiatry and Neuropsychiatric Genetics Research Group, Trinity Translational Medicine Institute, Trinity College DublinDublinIreland
- Department of Psychiatry, Medical Sciences Division, University of OxfordOxfordUnited Kingdom
- Department of Psychiatry, University Medical Center UtrechtUtrechtNetherlands
- Department of Medical Epidemiology and Biostatistics, Karolinska InstitutetStockholmSweden
- Department of Medical Epidemiology and Biostatistics Sweden, Karolinska InstitutetStockholmSweden
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm Health Care ServicesStockholmSweden
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrechtNetherlands
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Institute for Molecular Medicine FIMM, University of HelsinkiHelsinkiFinland
- Department of Public Health, University of HelsinkiHelsinkiFinland
- Faculty of Health, Medicine and Life Sciences, Maastricht UniversityMaastrichtNetherlands
- College of Pharmacy, University of ManitobaWinnipegCanada
- Centre for Neuroimaging and Cognitive Genomics (NICOG), School of Medicine, National University of Ireland GalwayGalwayIreland
- Division of Psychiatry, University College LondonLondonUnited Kingdom
- Department of Psychiatry, Royal College of Surgeons in IrelandDublinIreland
- Division of Biomedical Sciences, Institute of Health Research and Innovation, University of the Highlands and IslandsInvernessUnited Kingdom
- Department of Psychiatry and Psychotherapy, Jena University HospitalJenaGermany
- Department of Psychiatry and Psychotherapy, Philipps University Marburg/ Marburg University Hospital UKGMMarburgGermany
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht UniversityMaastrichtNetherlands
- The Institute of Medical Sciences, Univeristy of AberdeenAberdeenUnited Kingdom
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and WelfareHelsinkiFinland
- Department of Psychology and National Magnetic Resonance Research Center (UMRAM), Aysel Sabuncu Brain Research Centre (ASBAM), Bilkent UniversityAnkaraTurkey
- Molecular and Cellular Therapeutics, Royal College of Surgeons in IrelandDublinIreland
- Departments of Genetics and Psychiatry, University of North Carolina at Chapel HillChapel HillUnited States
- Neuroscience Genetics, Eli Lilly and CompanySurreyUnited Kingdom
- Department of Psychosis Studies, Institute of Psychiatry, King’s College LondonLondonUnited Kingdom
- School of Life Sciences, University of EssexColchesterUnited Kingdom
| | | | - Patrick Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska InstitutetStockholmSweden
- Departments of Genetics and Psychiatry, University of North Carolina at Chapel HillChapel HillUnited States
| | - Evangelos Vassos
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College LondonLondonUnited Kingdom
- NIHR BioResource Centre Maudsley, South London and Maudsley NHS Foundation Trust (SLaM), King’s College LondonLondonUnited Kingdom
| | | | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, King’s College LondonLondonUnited Kingdom
| | | | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, Barrack RoadExeterUnited Kingdom
| |
Collapse
|
11
|
Abstract
Schizophrenia is a serious neuropsychiatric disorder with abnormal age-related neurodevelopmental (or neurodegenerative) trajectories. Although an accelerated aging hypothesis of schizophrenia has been proposed, the quantitative study of the disruption of the physiological trajectory caused by schizophrenia is inconclusive. In this study, we employed 3 "epigenetic clock" methods to quantify the epigenetic age of a large sample size of whole blood (1069 samples from patients with schizophrenia vs 1264 samples from unaffected controls) and brain tissues (500 samples from patients with schizophrenia vs 711 samples from unaffected controls). We observed significant positive correlations between epigenetic age and chronological age in both blood and brain tissues from unaffected controls and patients with schizophrenia, as estimated by 3 methods. Furthermore, we observed that epigenetic age acceleration was significantly delayed in schizophrenia from the whole blood samples (aged 20-90 years) and brain frontal cortex tissues (aged 20-39 years). Intriguingly, the genes regulated by the epigenetic clock also contained schizophrenia-associated genes, displaying differential expression and methylation in patients with schizophrenia and involving in the regulation of cell activation and development. These findings were further supported by the dysregulated leukocyte composition in patients with schizophrenia. Our study presents quantitative evidence for a neurodevelopmental model of schizophrenia from the perspective of a skewed "epigenetic clock." Moreover, landmark changes in an easily accessible biological sample, blood, reveal the value of these epigenetic clock genes as peripheral biomarkers for schizophrenia.
Collapse
Affiliation(s)
- Xiaohui Wu
- Department of Medical Genetics, School of Basic Medical Sciences, Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, and Guangdong Engineering and Technology Research Center for Genetic Testing, Southern Medical University, Guangzhou, Guangdong, China,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, and Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, Guangdong, China
| | - Junping; Ye
- Department of Medical Genetics, School of Basic Medical Sciences, Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, and Guangdong Engineering and Technology Research Center for Genetic Testing, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongju Wang
- Department of Medical Genetics, School of Basic Medical Sciences, Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, and Guangdong Engineering and Technology Research Center for Genetic Testing, Southern Medical University, Guangzhou, Guangdong, China,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, and Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, Guangdong, China
| | - Cunyou Zhao
- Department of Medical Genetics, School of Basic Medical Sciences, Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, and Guangdong Engineering and Technology Research Center for Genetic Testing, Southern Medical University, Guangzhou, Guangdong, China,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, and Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, Guangdong, China,To whom correspondence should be addressed; Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; tel: (8620)-62789519, fax: (8620)-62789519, e-mail:
| |
Collapse
|
12
|
Smigielski L, Jagannath V, Rössler W, Walitza S, Grünblatt E. Epigenetic mechanisms in schizophrenia and other psychotic disorders: a systematic review of empirical human findings. Mol Psychiatry 2020; 25:1718-1748. [PMID: 31907379 DOI: 10.1038/s41380-019-0601-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 12/26/2022]
Abstract
Schizophrenia and other psychotic disorders are highly debilitating psychiatric conditions that lack a clear etiology and exhibit polygenic inheritance underlain by pleiotropic genes. The prevailing explanation points to the interplay between predisposing genes and environmental exposure. Accumulated evidence suggests that epigenetic regulation of the genome may mediate dynamic gene-environment interactions at the molecular level by modulating the expression of psychiatric phenotypes through transcription factors. This systematic review summarizes the current knowledge linking schizophrenia and other psychotic disorders to epigenetics, based on PubMed and Web of Science database searches conducted according to the PRISMA guidelines. Three groups of mechanisms in case-control studies of human tissue (i.e., postmortem brain and bio-fluids) were considered: DNA methylation, histone modifications, and non-coding miRNAs. From the initial pool of 3,204 records, 152 studies met our inclusion criteria (11,815/11,528, 233/219, and 2,091/1,827 cases/controls for each group, respectively). Many of the findings revealed associations with epigenetic modulations of genes regulating neurotransmission, neurodevelopment, and immune function, as well as differential miRNA expression (e.g., upregulated miR-34a, miR-7, and miR-181b). Overall, actual evidence moderately supports an association between epigenetics and schizophrenia and other psychotic disorders. However, heterogeneous results and cross-tissue extrapolations call for future work. Integrating epigenetics into systems biology may critically enhance research on psychosis and thus our understanding of the disorder. This may have implications for psychiatry in risk stratification, early recognition, diagnostics, precision medicine, and other interventional approaches targeting epigenetic fingerprints.
Collapse
Affiliation(s)
- Lukasz Smigielski
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland. .,The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.
| | - Vinita Jagannath
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.,Merck Sharp & Dohme (MSD) R&D Innovation Centre, London, UK
| | - Wulf Rössler
- The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.,Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland.,Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany.,Laboratory of Neuroscience, Institute of Psychiatry, Universidade de São Paulo, São Paulo, Brazil
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.,The Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University Hospital of Psychiatry Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Koshiyama D, Miura K, Nemoto K, Okada N, Matsumoto J, Fukunaga M, Hashimoto R. Neuroimaging studies within Cognitive Genetics Collaborative Research Organization aiming to replicate and extend works of ENIGMA. Hum Brain Mapp 2020; 43:182-193. [PMID: 32501580 PMCID: PMC8675417 DOI: 10.1002/hbm.25040] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/10/2020] [Accepted: 05/10/2020] [Indexed: 12/13/2022] Open
Abstract
Reproducibility is one of the most important issues for generalizing the results of clinical research; however, low reproducibility in neuroimaging studies is well known. To overcome this problem, the Enhancing Neuroimaging Genetics through Meta‐Analysis (ENIGMA) consortium, an international neuroimaging consortium, established standard protocols for imaging analysis and employs either meta‐ and mega‐analyses of psychiatric disorders with large sample sizes. The Cognitive Genetics Collaborative Research Organization (COCORO) in Japan promotes neurobiological studies in psychiatry and has successfully replicated and extended works of ENIGMA especially for neuroimaging studies. For example, (a) the ENIGMA consortium showed subcortical regional volume alterations in patients with schizophrenia (n = 2,028) compared to controls (n = 2,540) across 15 cohorts using meta‐analysis. COCORO replicated the volumetric changes in patients with schizophrenia (n = 884) compared to controls (n = 1,680) using the ENIGMA imaging analysis protocol and mega‐analysis. Furthermore, a schizophrenia‐specific leftward asymmetry for the pallidum volume was demonstrated; and (b) the ENIGMA consortium identified white matter microstructural alterations in patients with schizophrenia (n = 1,963) compared to controls (n = 2,359) across 29 cohorts. Using the ENIGMA protocol, a study from COCORO showed similar results in patients with schizophrenia (n = 696) compared to controls (n = 1,506) from 12 sites using mega‐analysis. Moreover, the COCORO study found that schizophrenia, bipolar disorder (n = 211) and autism spectrum disorder (n = 126), but not major depressive disorder (n = 398), share similar white matter microstructural alterations, compared to controls. Further replication and harmonization of the ENIGMA consortium and COCORO will contribute to the generalization of their research findings.
Collapse
Affiliation(s)
- Daisuke Koshiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenichiro Miura
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kiyotaka Nemoto
- Department of Psychiatry, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naohiro Okada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| | - Junya Matsumoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Masaki Fukunaga
- Division of Cerebral Integration, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
14
|
Wu Y, Li X, Liu J, Luo XJ, Yao YG. SZDB2.0: an updated comprehensive resource for schizophrenia research. Hum Genet 2020; 139:1285-1297. [DOI: 10.1007/s00439-020-02171-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/25/2020] [Indexed: 12/11/2022]
|
15
|
García-Gutiérrez MS, Navarrete F, Sala F, Gasparyan A, Austrich-Olivares A, Manzanares J. Biomarkers in Psychiatry: Concept, Definition, Types and Relevance to the Clinical Reality. Front Psychiatry 2020; 11:432. [PMID: 32499729 PMCID: PMC7243207 DOI: 10.3389/fpsyt.2020.00432] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
During the last years, an extraordinary effort has been made to identify biomarkers as potential tools for improving prevention, diagnosis, drug response and drug development in psychiatric disorders. Contrary to other diseases, mental illnesses are classified by diagnostic categories with a broad variety list of symptoms. Consequently, patients diagnosed from the same psychiatric illness present a great heterogeneity in their clinical presentation. This fact together with the incomplete knowledge of the neurochemical alterations underlying mental disorders, contribute to the limited efficacy of current pharmacological options. In this respect, the identification of biomarkers in psychiatry is becoming essential to facilitate diagnosis through the developing of markers that allow to stratify groups within the syndrome, which in turn may lead to more focused treatment options. In order to shed light on this issue, this review summarizes the concept and types of biomarkers including an operational definition for therapeutic development. Besides, the advances in this field were summarized and sorted into five categories, which include genetics, transcriptomics, proteomics, metabolomics, and epigenetics. While promising results were achieved, there is a lack of biomarker investigations especially related to treatment response to psychiatric conditions. This review includes a final conclusion remarking the future challenges required to reach the goal of developing valid, reliable and broadly-usable biomarkers for psychiatric disorders and their treatment. The identification of factors predicting treatment response will reduce trial-and-error switches of medications facilitating the discovery of new effective treatments, being a crucial step towards the establishment of greater personalized medicine.
Collapse
Affiliation(s)
- Maria Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Francisco Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | | | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| |
Collapse
|
16
|
Jansen RJ, Tong L, Argos M, Jasmine F, Rakibuz-Zaman M, Sarwar G, Islam MT, Shahriar H, Islam T, Rahman M, Yunus M, Kibriya MG, Baron JA, Ahsan H, Pierce BL. The effect of age on DNA methylation in whole blood among Bangladeshi men and women. BMC Genomics 2019; 20:704. [PMID: 31506065 PMCID: PMC6734473 DOI: 10.1186/s12864-019-6039-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/16/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND It is well-known that methylation changes occur as humans age, however, understanding how age-related changes in DNA methylation vary by sex is lacking. In this study, we characterize the effect of age on DNA methylation in a sex-specific manner and determine if these effects vary by genomic context. We used the Illumina HumanMethylation 450 K array and DNA derived from whole blood for 400 adult participants (189 males and 211 females) from Bangladesh to identify age-associated CpG sites and regions and characterize the location of these age-associated sites with respect to CpG islands (vs. shore, shelf, or open sea) and gene regions (vs. intergenic). We conducted a genome-wide search for age-associated CpG sites (among 423,604 sites) using a reference-free approach to adjust for cell type composition (the R package RefFreeEWAS) and performed an independent replication analysis of age-associated CpGs. RESULTS The number of age-associated CpGs (p < 5 x 10- 8) were 986 among men and 3479 among women of which 2027(63.8%) and 572 (64.1%) replicated (using Bonferroni adjusted p < 1.2 × 10- 5). For both sexes, age-associated CpG sites were more likely to be hyper-methylated with increasing age (compared to hypo-methylated) and were enriched in CpG islands and promoter regions compared with other locations and all CpGs on the array. Although we observed strong correlation between chronological age and previously-developed epigenetic age models (r ≈ 0.8), among our top (based on lowest p-value) age-associated CpG sites only 12 for males and 44 for females are included in these prediction models, and the median chronological age compared to predicted age was 44 vs. 51.7 in males and 45 vs. 52.1 in females. CONCLUSIONS Our results describe genome-wide features of age-related changes in DNA methylation. The observed associations between age and methylation were generally consistent for both sexes, although the associations tended to be stronger among women. Our population may have unique age-related methylation changes that are not captured in the established methylation-based age prediction model we used, which was developed to be non-tissue-specific.
Collapse
Affiliation(s)
- Rick J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
- Biostatistics Core Facility, North Dakota State University, Fargo, ND, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, 5841 S. Maryland Ave., W264, MC2000, Chicago, IL, 60637, USA
| | - Maria Argos
- Divison of Epidemiology and Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, IL, USA
| | - Farzana Jasmine
- Department of Public Health Sciences, University of Chicago, 5841 S. Maryland Ave., W264, MC2000, Chicago, IL, 60637, USA
| | | | - Golam Sarwar
- UChicago Research Bangladesh Mohakhali, Dhaka, 1230, Bangladesh
| | | | - Hasan Shahriar
- UChicago Research Bangladesh Mohakhali, Dhaka, 1230, Bangladesh
| | - Tariqul Islam
- UChicago Research Bangladesh Mohakhali, Dhaka, 1230, Bangladesh
| | - Mahfuzar Rahman
- UChicago Research Bangladesh Mohakhali, Dhaka, 1230, Bangladesh
- Research and Evaluation Division BRAC, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Yunus
- International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, 1000, Bangladesh
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, 5841 S. Maryland Ave., W264, MC2000, Chicago, IL, 60637, USA
| | - John A Baron
- Department of Epidemiology, Gillings School of Global Public Health, University of North Caroline, Chapel Hill, NC, USA
| | - Habibul Ahsan
- Department of Public Health Sciences, University of Chicago, 5841 S. Maryland Ave., W264, MC2000, Chicago, IL, 60637, USA.
- Department of Medicine, The University of Chicago, Chicago, IL, USA.
- Department of Human Genetics and Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA.
| | - Brandon L Pierce
- Department of Public Health Sciences, University of Chicago, 5841 S. Maryland Ave., W264, MC2000, Chicago, IL, 60637, USA.
- Department of Human Genetics and Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
17
|
Kong Y, Rastogi D, Seoighe C, Greally JM, Suzuki M. Insights from deconvolution of cell subtype proportions enhance the interpretation of functional genomic data. PLoS One 2019; 14:e0215987. [PMID: 31022271 PMCID: PMC6483354 DOI: 10.1371/journal.pone.0215987] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/11/2019] [Indexed: 02/07/2023] Open
Abstract
Cell subtype proportion variability between samples contributes significantly to the variation of functional genomic properties such as gene expression or DNA methylation. Although the impact of the variation of cell subtype composition on measured genomic quantities is recognized, and some innovative tools have been developed for the analysis of heterogeneous samples, most functional genomics studies using samples with mixed cell types still ignore the influence of cell subtype proportion variation, or just deal with it as a nuisance variable to be eliminated. Here we demonstrate how harvesting information about cell subtype proportions from functional genomics data can provide insights into cellular changes associated with phenotypes. We focused on two types of mixed cell populations, human blood and mouse kidney. Cell type prediction is well developed in the former, but not currently in the latter. Estimating the cellular repertoire is easier when a reference dataset from purified samples of all cell types in the tissue is available, as is the case for blood. However, reference datasets are not available for most other tissues, such as the kidney. In this study, we showed that the proportion of alterations attributable to changes in the cellular composition varies strikingly in the two disorders (asthma and systemic lupus erythematosus), suggesting that the contribution of cell subtype proportion changes to functional genomic properties can be disease-specific. We also showed that a reference dataset from a single-cell RNA-seq study successfully estimated the cell subtype proportions in mouse kidney and allowed us to distinguish altered cell subtype differences between two different knock-out mouse models, both of which had reported a reduced number of glomeruli compared to their wild-type counterparts. These findings demonstrate that testing for changes in cell subtype proportions between conditions can yield important insights in functional genomics studies.
Collapse
Affiliation(s)
- Yu Kong
- Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Deepa Rastogi
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Cathal Seoighe
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland Galway, University Road, Galway, Ireland
| | - John M. Greally
- Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Masako Suzuki
- Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
18
|
Liu C, Jiao C, Wang K, Yuan N. DNA Methylation and Psychiatric Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:175-232. [PMID: 29933950 DOI: 10.1016/bs.pmbts.2018.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
DNA methylation has been an important area of research in the study of molecular mechanism to psychiatric disorders. Recent evidence has suggested that abnormalities in global methylation, methylation of genes, and pathways could play a role in the etiology of many forms of mental illness. In this article, we review the mechanisms of DNA methylation, including the genetic and environmental factors affecting methylation changes. We report and discuss major findings regarding DNA methylation in psychiatric patients, both within the context of global methylation studies and gene-specific methylation studies. Finally, we discuss issues surrounding data quality improvement, the limitations of current methylation analysis methods, and the possibility of using DNA methylation-based treatment for psychiatric disorders in the future.
Collapse
Affiliation(s)
- Chunyu Liu
- University of Illinois, Chicago, IL, United States; School of Life Science, Central South University, Changsha, China.
| | - Chuan Jiao
- School of Life Science, Central South University, Changsha, China
| | - Kangli Wang
- School of Life Science, Central South University, Changsha, China
| | - Ning Yuan
- Hunan Brain Hospital, Changsha, China
| |
Collapse
|
19
|
Tomioka Y, Numata S, Kinoshita M, Umehara H, Watanabe SY, Nakataki M, Iwayama Y, Toyota T, Ikeda M, Yamamori H, Shimodera S, Tajima A, Hashimoto R, Iwata N, Yoshikawa T, Ohmori T. Decreased serum pyridoxal levels in schizophrenia: meta-analysis and Mendelian randomization analysis. J Psychiatry Neurosci 2018; 43:170053. [PMID: 29402374 PMCID: PMC5915240 DOI: 10.1503/jpn.170053] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/21/2017] [Accepted: 10/22/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alterations in one-carbon metabolism have been associated with schizophrenia, and vitamin B6 is one of the key components in this pathway. METHODS We first conducted a case-control study of serum pyridoxal levels and schizophrenia in a large Japanese cohort (n = 1276). Subsequently, we conducted a meta-analysis of association studies (n = 2125). Second, we investigated whether rs4654748, which was identified in a genome-wide association study as a vitamin B6-related single nucleotide polymorphism, was genetically implicated in patients with schizophrenia in the Japanese population (n = 10 689). Finally, we assessed the effect of serum pyridoxal levels on schizophrenia risk using a Mendelian randomization (MR) approach. RESULTS Serum pyridoxal levels were significantly lower in patients with schizophrenia than in controls, not only in our cohort, but also in the pooled data set of the meta-analysis of association studies (standardized mean difference -0.48, 95% confidence interval [CI] -0.57 to -0.39, p = 9.8 × 10-24). We failed to find a significant association between rs4654748 and schizophrenia. Furthermore, an MR analysis failed to find a causal relationship between pyridoxal levels and schizophrenia risk (odds ratio 0.99, 95% CI 0.65-1.51, p = 0.96). LIMITATIONS Food consumption and medications may have affected serum pyridoxal levels in our cross-sectional study. Sample size, number of instrumental variables and substantial heterogeneity among patients with schizophrenia are limitations of an MR analysis. CONCLUSION We found decreased serum pyridoxal levels in patients with schizophrenia in this observational study. However, we failed to obtain data supporting a causal relationship between pyridoxal levels and schizophrenia risk using the MR approach.
Collapse
Affiliation(s)
- Yukiko Tomioka
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Shusuke Numata
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Makoto Kinoshita
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Hidehiro Umehara
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Shin-Ya Watanabe
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Masahito Nakataki
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Yoshimi Iwayama
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Tomoko Toyota
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Masashi Ikeda
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Hidenaga Yamamori
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Shinji Shimodera
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Atsushi Tajima
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Ryota Hashimoto
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Nakao Iwata
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Takeo Yoshikawa
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| | - Tetsuro Ohmori
- From the Department of Psychiatry, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan (Tomioka, Numata, Kinoshita, Umehara, Watanabe, Nakataki, Ohmori); the Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan (Iwayama, Toyota, Yoshikawa); the Department of Psychiatry, School of Medicine, Fujita Health University, Aichi, Japan (Ikeda, Iwata); the Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan (Yamamori, Hashimoto); the Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan (Shimodera); the Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa, Japan (Tajima); and the Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan (Hashimoto)
| |
Collapse
|
20
|
Variation in global DNA hydroxymethylation with age associated with schizophrenia. Psychiatry Res 2017; 257:497-500. [PMID: 28841512 DOI: 10.1016/j.psychres.2017.08.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/08/2017] [Accepted: 08/12/2017] [Indexed: 12/31/2022]
Abstract
To improve understanding of DNA hydroxymethylation (5hmC) and methylation (5mC) in the development of schizophrenia, this study examined global 5hmC and 5mC levels in peripheral blood DNA of 264 patients with schizophrenia and 221 controls and observed increased 5mC levels in the patients and increased 5hmC levels in male patients but decreased levels in female patients as compared with the controls. The 5mC level displayed a gender-dependent positive correlation with age and the 5hmC level displayed a correlation with age positively in controls but negatively in patients, and their role in the pathogenesis of schizophrenia remains to be elucidated.
Collapse
|
21
|
Saliva as a Blood Alternative for Genome-Wide DNA Methylation Profiling by Methylated DNA Immunoprecipitation (MeDIP) Sequencing. EPIGENOMES 2017. [DOI: 10.3390/epigenomes1030014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
22
|
Schmitt A, Martins-de-Souza D, Akbarian S, Cassoli JS, Ehrenreich H, Fischer A, Fonteh A, Gattaz WF, Gawlik M, Gerlach M, Grünblatt E, Halene T, Hasan A, Hashimoto K, Kim YK, Kirchner SK, Kornhuber J, Kraus TFJ, Malchow B, Nascimento JM, Rossner M, Schwarz M, Steiner J, Talib L, Thibaut F, Riederer P, Falkai P. Consensus paper of the WFSBP Task Force on Biological Markers: Criteria for biomarkers and endophenotypes of schizophrenia, part III: Molecular mechanisms. World J Biol Psychiatry 2017; 18:330-356. [PMID: 27782767 DOI: 10.1080/15622975.2016.1224929] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Despite progress in identifying molecular pathophysiological processes in schizophrenia, valid biomarkers are lacking for both the disease and treatment response. METHODS This comprehensive review summarises recent efforts to identify molecular mechanisms on the level of protein and gene expression and epigenetics, including DNA methylation, histone modifications and micro RNA expression. Furthermore, it summarises recent findings of alterations in lipid mediators and highlights inflammatory processes. The potential that this research will identify biomarkers of schizophrenia is discussed. RESULTS Recent studies have not identified clear biomarkers for schizophrenia. Although several molecular pathways have emerged as potential candidates for future research, a complete understanding of these metabolic pathways is required to reveal better treatment modalities for this disabling condition. CONCLUSIONS Large longitudinal cohort studies are essential that pair a thorough phenotypic and clinical evaluation for example with gene expression and proteome analysis in blood at multiple time points. This approach might identify biomarkers that allow patients to be stratified according to treatment response and ideally also allow treatment response to be predicted. Improved knowledge of molecular pathways and epigenetic mechanisms, including their potential association with environmental influences, will facilitate the discovery of biomarkers that could ultimately be effective tools in clinical practice.
Collapse
Affiliation(s)
- Andrea Schmitt
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany.,b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil
| | - Daniel Martins-de-Souza
- b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil.,c Laboratory of Neuroproteomics, Department of Biochemistry , Institute of Biology University of Campinas (UNICAMP), Campinas , SP , Brazil
| | - Schahram Akbarian
- d Division of Psychiatric Epigenomics, Departments of Psychiatry and Neuroscience , Mount Sinai School of Medicine , New York , USA
| | - Juliana S Cassoli
- c Laboratory of Neuroproteomics, Department of Biochemistry , Institute of Biology University of Campinas (UNICAMP), Campinas , SP , Brazil
| | - Hannelore Ehrenreich
- e Clinical Neuroscience , Max Planck Institute of Experimental Medicine, DFG Centre for Nanoscale Microscopy & Molecular Physiology of the Brain , Göttingen , Germany
| | - Andre Fischer
- f Research Group for Epigenetics in Neurodegenerative Diseases , German Centre for Neurodegenerative Diseases (DZNE), Göttingen , Germany.,g Department of Psychiatry and Psychotherapy , University Medical Centre Göttingen , Germany
| | - Alfred Fonteh
- h Neurosciences , Huntington Medical Research Institutes , Pasadena , CA , USA
| | - Wagner F Gattaz
- b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil
| | - Michael Gawlik
- i Department of Psychiatry and Psychotherapy , University of Würzburg , Germany
| | - Manfred Gerlach
- j Centre for Mental Health, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University of Würzburg , Germany
| | - Edna Grünblatt
- i Department of Psychiatry and Psychotherapy , University of Würzburg , Germany.,k Department of Child and Adolescent Psychiatry and Psychotherapy , Psychiatric Hospital, University of Zürich , Switzerland.,l Neuroscience Centre Zurich , University of Zurich and the ETH Zurich , Switzerland.,m Zurich Centre for Integrative Human Physiology , University of Zurich , Switzerland
| | - Tobias Halene
- d Division of Psychiatric Epigenomics, Departments of Psychiatry and Neuroscience , Mount Sinai School of Medicine , New York , USA
| | - Alkomiet Hasan
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany
| | - Kenij Hashimoto
- n Division of Clinical Neuroscience , Chiba University Centre for Forensic Mental Health , Chiba , Japan
| | - Yong-Ku Kim
- o Department of Psychiatry , Korea University, College of Medicine , Republic of Korea
| | | | - Johannes Kornhuber
- p Department of Psychiatry and Psychotherapy , Friedrich-Alexander-University Erlangen-Nuremberg , Erlangen , Germany
| | | | - Berend Malchow
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany
| | - Juliana M Nascimento
- c Laboratory of Neuroproteomics, Department of Biochemistry , Institute of Biology University of Campinas (UNICAMP), Campinas , SP , Brazil
| | - Moritz Rossner
- r Department of Psychiatry, Molecular and Behavioural Neurobiology , LMU Munich , Germany.,s Research Group Gene Expression , Max Planck Institute of Experimental Medicine , Göttingen , Germany
| | - Markus Schwarz
- t Institute for Laboratory Medicine, LMU Munich , Germany
| | - Johann Steiner
- u Department of Psychiatry , University of Magdeburg , Magdeburg , Germany
| | - Leda Talib
- b Laboratory of Neuroscience (LIM27) , Institute of Psychiatry, University of Sao Paulo , Sao Paulo , Brazil
| | - Florence Thibaut
- v Department of Psychiatry , University Hospital Cochin (site Tarnier), University of Paris-Descartes, INSERM U 894 Centre Psychiatry and Neurosciences , Paris , France
| | - Peter Riederer
- w Center of Psychic Health; Department of Psychiatry, Psychosomatics and Psychotherapy , University Hospital of Würzburg , Germany
| | - Peter Falkai
- a Department of Psychiatry and Psychotherapy , LMU Munich , Germany
| | | |
Collapse
|
23
|
Zong L, Zhou L, Hou Y, Zhang L, Jiang W, Zhang W, Wang L, Luo X, Wang S, Deng C, Peng Z, Li S, Hu J, Zhao H, Zhao C. Genetic and epigenetic regulation on the transcription of GABRB2: Genotype-dependent hydroxymethylation and methylation alterations in schizophrenia. J Psychiatr Res 2017; 88:9-17. [PMID: 28063323 DOI: 10.1016/j.jpsychires.2016.12.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/30/2016] [Accepted: 12/19/2016] [Indexed: 12/17/2022]
Abstract
To improve our understanding of the abnormalities and non-Mendelian inheritance characteristics of schizophrenia, this study examined DNA methylation (5mC) and hydroxymethylation (5hmC) in the schizophrenia-associated GABRB2 gene encoding the type A γ-aminobutyric acid receptor β2 subunit. DNAs from the peripheral white blood cells of 279 schizophrenic patients and 256 controls from the Chinese Han population were examined to reveal that the GABRB2 promoter P1-5mC level which was correlated with olanzapine administration, P2-5mC/5hmC level, and Alu-5mC level which was correlated with administration of ziprasidone or oxcarbazepine, were increased in schizophrenic patients. Significant correlations of the promoter 5mC/5hmC levels with the genotypes of single nucleotide polymorphisms (SNPs) were observed with SNPs rs72815526 (C/A) and rs3811997 (C/T). In schizophrenics, the heterozygous genotypes of rs72815526 (C/A) were correlated with increased 5hmC levels whereas the heterozygous genotypes of rs3811997 (C/T) were correlated with decreased 5mC levels. Moreover, the GABRB2 promoter with rs3811997(C/T) minor allele T or the methylation-disrupted type AG at -254 and -231 CCGG sites was observed to enhance the promoter activity in the luciferase reporter-transfected human embryonic kidney 293 cells. An elevated GABRB2 mRNA transcriptional level in human neuroblastoma IMR32 cells were accompanied by the decreased promoter 5hmC/5mC levels induced by 5-azacytidine or by increased histone H4 acetylation levels of the Alu-Yi6 region induced by valproic acid. These results reveal alterations in GABRB2 genotype-dependent methylation and hydroxymethylation in schizophrenia, which yielded transcriptional and translational alterations in the cultured cells, and help elucidate the genetic-epigenetic interactions influencing schizophrenia.
Collapse
Affiliation(s)
- Lu Zong
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China
| | - Lin Zhou
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China; Key Laboratory of Genetics and Birth Health of Hunan Province, Family Planning Institute of Hunan Province, Changsha, China
| | - Yu Hou
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China
| | - Lulu Zhang
- Department of Psychiatry, Guangzhou First People's Hospital, Guangzhou, Guangdong, China
| | - Wei Jiang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China
| | - Wenwei Zhang
- The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Lijuan Wang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China
| | - Xia Luo
- Department of Psychiatry, Shenzhen Kangning Hospital & Shenzhen Mental Health Center, Shenzhen, China
| | - Shiqing Wang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China
| | - Cong Deng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhizhen Peng
- The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Shufen Li
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China
| | - Jiming Hu
- The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Hu Zhao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cunyou Zhao
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Technology and Engineering Research Center for Molecular Diagnostics of Human Genetic Diseases, Guangzhou, Guangdong, China.
| |
Collapse
|
24
|
Effect of Clozapine on DNA Methylation in Peripheral Leukocytes from Patients with Treatment-Resistant Schizophrenia. Int J Mol Sci 2017; 18:ijms18030632. [PMID: 28335437 PMCID: PMC5372645 DOI: 10.3390/ijms18030632] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 02/06/2023] Open
Abstract
Clozapine is an atypical antipsychotic, that is established as the treatment of choice for treatment-resistant schizophrenia (SCZ). To date, no study investigating comprehensive DNA methylation changes in SCZ patients treated with chronic clozapine has been reported. The purpose of the present study is to reveal the effects of clozapine on DNA methylation in treatment-resistant SCZ. We conducted a genome-wide DNA methylation profiling in peripheral leukocytes (485,764 CpG dinucleotides) from treatment-resistant SCZ patients treated with clozapine (n = 21) in a longitudinal study. Significant changes in DNA methylation were observed at 29,134 sites after one year of treatment with clozapine, and these genes were enriched for “cell substrate adhesion” and “cell matrix adhesion” gene ontology (GO) terms. Furthermore, DNA methylation changes in the CREBBP (CREB binding protein) gene were significantly correlated with the clinical improvements. Our findings provide insights into the action of clozapine in treatment-resistant SCZ.
Collapse
|
25
|
Ji H, Wang Y, Liu G, Chang L, Chen Z, Zhou D, Xu X, Cui W, Hong Q, Jiang L, Li J, Zhou X, Li Y, Guo Z, Zha Q, Niu Y, Weng Q, Duan S, Wang Q. Elevated OPRD1 promoter methylation in Alzheimer's disease patients. PLoS One 2017; 12:e0172335. [PMID: 28253273 PMCID: PMC5333823 DOI: 10.1371/journal.pone.0172335] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/03/2017] [Indexed: 01/21/2023] Open
Abstract
Aberrant DNA methylation has been observed in the patients with Alzheimer’s disease (AD), a common neurodegenerative disorder in the elderly. OPRD1 encodes the delta opioid receptor, a member of the opioid family of G-protein-coupled receptors. In the current study, we compare the DNA methylation levels of OPRD1 promoter CpG sites (CpG1, CpG2, and CpG3) between 51 AD cases and 63 controls using the bisulfite pyrosequencing technology. Our results show that significantly higher CpG3 methylation is found in AD cases than controls. Significant associations are found between several biochemical parameters (including HDL-C and ALP) and CpG3 methylation. Subsequent luciferase reporter gene assay shows that DNA fragment containing the three OPRD1 promoter CpGs is able to regulate gene expression. In summary, our results suggest that OPRD1 promoter hypermethylation is associated with the risk of AD.
Collapse
Affiliation(s)
- Huihui Ji
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yunliang Wang
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, Shandong, China
| | - Guili Liu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Lan Chang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | | | | | - Xuting Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Wei Cui
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Qingxiao Hong
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Liting Jiang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Jinfeng Li
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, Shandong, China
| | - Xiaohui Zhou
- Department of Internal Medicine for Cadres, the First Affiliated Hospital of Xinjiang Medical University, Urumchi, China
| | - Ying Li
- Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
| | - Zhiping Guo
- School of Medicine, Lishui University, Lishui, Zhejiang, China
| | - Qin Zha
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (QW); (SD); (QZ)
| | - Yanfang Niu
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qiuyan Weng
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shiwei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (QW); (SD); (QZ)
| | - Qinwen Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (QW); (SD); (QZ)
| |
Collapse
|
26
|
Epigenome-wide association study of DNA methylation in panic disorder. Clin Epigenetics 2017; 9:6. [PMID: 28149334 PMCID: PMC5270210 DOI: 10.1186/s13148-016-0307-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/26/2016] [Indexed: 12/22/2022] Open
Abstract
Background Panic disorder (PD) is considered to be a multifactorial disorder emerging from interactions among multiple genetic and environmental factors. To date, although genetic studies reported several susceptibility genes with PD, few of them were replicated and the pathogenesis of PD remains to be clarified. Epigenetics is considered to play an important role in etiology of complex traits and diseases, and DNA methylation is one of the major forms of epigenetic modifications. In this study, we performed an epigenome-wide association study of PD using DNA methylation arrays so as to investigate the possibility that different levels of DNA methylation might be associated with PD. Methods The DNA methylation levels of CpG sites across the genome were examined with genomic DNA samples (PD, N = 48, control, N = 48) extracted from peripheral blood. Methylation arrays were used for the analysis. β values, which represent the levels of DNA methylation, were normalized via an appropriate pipeline. Then, β values were converted to M values via the logit transformation for epigenome-wide association study. The relationship between each DNA methylation site and PD was assessed by linear regression analysis with adjustments for the effects of leukocyte subsets. Results Forty CpG sites showed significant association with PD at 5% FDR correction, though the differences of the DNA methylation levels were relatively small. Most of the significant CpG sites (37/40 CpG sites) were located in or around CpG islands. Many of the significant CpG sites (27/40 CpG sites) were located upstream of genes, and all such CpG sites with the exception of two were hypomethylated in PD subjects. A pathway analysis on the genes annotated to the significant CpG sites identified several pathways, including “positive regulation of lymphocyte activation.” Conclusions Although future studies with larger number of samples are necessary to confirm the small DNA methylation abnormalities associated with PD, there is a possibility that several CpG sites might be associated, together as a group, with PD. Electronic supplementary material The online version of this article (doi:10.1186/s13148-016-0307-1) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
Abstract
Schizophrenia is a highly heritable psychiatric condition that displays a complex phenotype. A multitude of genetic susceptibility loci have now been identified, but these fail to explain the high heritability estimates of schizophrenia. In addition, epidemiologically relevant environmental risk factors for schizophrenia may lead to permanent changes in brain function. In conjunction with genetic liability, these environmental risk factors-likely through epigenetic mechanisms-may give rise to schizophrenia, a clinical syndrome characterized by florid psychotic symptoms and moderate to severe cognitive impairment. These pathophysiological features point to the involvement of epigenetic processes. Recently, a wave of studies examining aberrant DNA modifications in schizophrenia was published. This chapter aims to comprehensively review the current findings, from both candidate gene studies and genome-wide approaches, on DNA methylation changes in schizophrenia.
Collapse
|
28
|
Hannon E, Dempster E, Viana J, Burrage J, Smith AR, Macdonald R, St Clair D, Mustard C, Breen G, Therman S, Kaprio J, Toulopoulou T, Pol HEH, Bohlken MM, Kahn RS, Nenadic I, Hultman CM, Murray RM, Collier DA, Bass N, Gurling H, McQuillin A, Schalkwyk L, Mill J. An integrated genetic-epigenetic analysis of schizophrenia: evidence for co-localization of genetic associations and differential DNA methylation. Genome Biol 2016; 17:176. [PMID: 27572077 PMCID: PMC5004279 DOI: 10.1186/s13059-016-1041-x] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/09/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Schizophrenia is a highly heritable, neuropsychiatric disorder characterized by episodic psychosis and altered cognitive function. Despite success in identifying genetic variants associated with schizophrenia, there remains uncertainty about the causal genes involved in disease pathogenesis and how their function is regulated. RESULTS We performed a multi-stage epigenome-wide association study, quantifying genome-wide patterns of DNA methylation in a total of 1714 individuals from three independent sample cohorts. We have identified multiple differentially methylated positions and regions consistently associated with schizophrenia across the three cohorts; these effects are independent of important confounders such as smoking. We also show that epigenetic variation at multiple loci across the genome contributes to the polygenic nature of schizophrenia. Finally, we show how DNA methylation quantitative trait loci in combination with Bayesian co-localization analyses can be used to annotate extended genomic regions nominated by studies of schizophrenia, and to identify potential regulatory variation causally involved in disease. CONCLUSIONS This study represents the first systematic integrated analysis of genetic and epigenetic variation in schizophrenia, introducing a methodological approach that can be used to inform epigenome-wide association study analyses of other complex traits and diseases. We demonstrate the utility of using a polygenic risk score to identify molecular variation associated with etiological variation, and of using DNA methylation quantitative trait loci to refine the functional and regulatory variation associated with schizophrenia risk variants. Finally, we present strong evidence for the co-localization of genetic associations for schizophrenia and differential DNA methylation.
Collapse
Affiliation(s)
- Eilis Hannon
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Emma Dempster
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Joana Viana
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Joe Burrage
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Adam R. Smith
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Ruby Macdonald
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - David St Clair
- The Institute of Medical Sciences, Aberdeen University, Aberdeen, UK
| | | | - Gerome Breen
- Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
| | | | - Jaakko Kaprio
- National Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | | | - Hilleke E. Hulshoff Pol
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marc M. Bohlken
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rene S. Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Igor Nenadic
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Christina M. Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - Robin M. Murray
- Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
| | - David A. Collier
- Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
- Eli Lilly and Company Ltd, Windlesham, UK
| | - Nick Bass
- Division of Psychiatry, University College London, London, UK
| | - Hugh Gurling
- Division of Psychiatry, University College London, London, UK
| | | | - Leonard Schalkwyk
- Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
- School of Biological Sciences, University of Essex, Colchester, UK
| | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, Exeter, UK
- Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King’s College London, London, UK
- Royal Devon & Exeter Hospital, RILD Building, Level 4, Barrack Rd, Exeter, EX2 5DW UK
| |
Collapse
|
29
|
Cariaga-Martinez A, Saiz-Ruiz J, Alelú-Paz R. From Linkage Studies to Epigenetics: What We Know and What We Need to Know in the Neurobiology of Schizophrenia. Front Neurosci 2016; 10:202. [PMID: 27242407 PMCID: PMC4862989 DOI: 10.3389/fnins.2016.00202] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/25/2016] [Indexed: 01/15/2023] Open
Abstract
Schizophrenia is a complex psychiatric disorder characterized by the presence of positive, negative, and cognitive symptoms that lacks a unifying neuropathology. In the present paper, we will review the current understanding of molecular dysregulation in schizophrenia, including genetic and epigenetic studies. In relation to the latter, basic research suggests that normal cognition is regulated by epigenetic mechanisms and its dysfunction occurs upon epigenetic misregulation, providing new insights into missing heritability of complex psychiatric diseases, referring to the discrepancy between epidemiological heritability and the proportion of phenotypic variation explained by DNA sequence difference. In schizophrenia the absence of consistently replicated genetic effects together with evidence for lasting changes in gene expression after environmental exposures suggest a role of epigenetic mechanisms. In this review we will focus on epigenetic modifications as a key mechanism through which environmental factors interact with individual's genetic constitution to affect risk of psychotic conditions throughout life.
Collapse
Affiliation(s)
- Ariel Cariaga-Martinez
- Laboratory for Neuroscience of Mental Disorders Elena Pessino, Department of Medicine and Medical Specialties, School of Medicine, Alcalá University Madrid, Spain
| | - Jerónimo Saiz-Ruiz
- Department of Psychiatry, Ramón y Cajal Hospital, IRYCISMadrid, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)Madrid, Spain
| | - Raúl Alelú-Paz
- Laboratory for Neuroscience of Mental Disorders Elena Pessino, Department of Medicine and Medical Specialties, School of Medicine, Alcalá UniversityMadrid, Spain; Department of Psychiatry, Ramón y Cajal Hospital, IRYCISMadrid, Spain
| |
Collapse
|
30
|
Montano C, Taub MA, Jaffe A, Briem E, Feinberg JI, Trygvadottir R, Idrizi A, Runarsson A, Berndsen B, Gur RC, Moore TM, Perry RT, Fugman D, Sabunciyan S, Yolken RH, Hyde TM, Kleinman JE, Sobell JL, Pato CN, Pato MT, Go RC, Nimgaonkar V, Weinberger DR, Braff D, Gur RE, Fallin MD, Feinberg AP. Association of DNA Methylation Differences With Schizophrenia in an Epigenome-Wide Association Study. JAMA Psychiatry 2016; 73:506-14. [PMID: 27074206 PMCID: PMC6353566 DOI: 10.1001/jamapsychiatry.2016.0144] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE DNA methylation may play an important role in schizophrenia (SZ), either directly as a mechanism of pathogenesis or as a biomarker of risk. OBJECTIVE To scan genome-wide DNA methylation data to identify differentially methylated CpGs between SZ cases and controls. DESIGN, SETTING, AND PARTICIPANTS Epigenome-wide association study begun in 2008 using DNA methylation levels of 456 513 CpG loci measured on the Infinium HumanMethylation450 array (Illumina) in a consortium of case-control studies for initial discovery and in an independent replication set. Primary analyses used general linear regression, adjusting for age, sex, race/ethnicity, smoking, batch, and cell type heterogeneity. The discovery set contained 689 SZ cases and 645 controls (n = 1334), from 3 multisite consortia: the Consortium on the Genetics of Endophenotypes in Schizophrenia, the Project among African-Americans To Explore Risks for Schizophrenia, and the Multiplex Multigenerational Family Study of Schizophrenia. The replication set contained 247 SZ cases and 250 controls (n = 497) from the Genomic Psychiatry Cohort. MAIN OUTCOMES AND MEASURES Identification of differentially methylated positions across the genome in SZ cases compared with controls. RESULTS Of the 689 case participants in the discovery set, 477 (69%) were men and 258 (37%) were non-African American; of the 645 controls, 273 (42%) were men and 419 (65%) were non-African American. In our replication set, cases/controls were 76% male and 100% non-African American. We identified SZ-associated methylation differences at 923 CpGs in the discovery set (false discovery rate, <0.2). Of these, 625 showed changes in the same direction including 172 with P < .05 in the replication set. Some replicated differentially methylated positions are located in a top-ranked SZ region from genome-wide association study analyses. CONCLUSIONS AND RELEVANCE This analysis identified 172 replicated new associations with SZ after careful correction for cell type heterogeneity and other potential confounders. The overlap with previous genome-wide association study data can provide potential insights into the functional relevance of genetic signals for SZ.
Collapse
Affiliation(s)
- Carolina Montano
- Medical Scientist Training Program and Predoctoral Training Program in Human Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland,Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Margaret A. Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Andrew Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Eirikur Briem
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jason I. Feinberg
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rakel Trygvadottir
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Adrian Idrizi
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Arni Runarsson
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Birna Berndsen
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ruben C. Gur
- Neuropsychiatry Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tyler M. Moore
- Neuropsychiatry Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rodney T. Perry
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham
| | - Doug Fugman
- Rutgers University Cell and DNA Repository, Piscataway, New Jersey
| | - Sarven Sabunciyan
- Stanley Division of Developmental Neurovirology, Johns Hopkins, School of Medicine, Baltimore, Maryland
| | - Robert H. Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins, School of Medicine, Baltimore, Maryland
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Janet L. Sobell
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine of University of Southern California, Los Angeles
| | - Carlos N. Pato
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine of University of Southern California, Los Angeles
| | - Michele T. Pato
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine of University of Southern California, Los Angeles
| | - Rodney C. Go
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham
| | | | - Daniel R. Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - David Braff
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla,VISN22, Mental Illness Research, Education, and Clinical Center, VA Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Raquel E. Gur
- Neuropsychiatry Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Margaret Daniele Fallin
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Andrew P. Feinberg
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland,Departments of Medicine and Biomedical Engineering, Johns Hopkins University School of Medicine and Whiting School of Engineering, Baltimore, Maryland
| |
Collapse
|
31
|
Teroganova N, Girshkin L, Suter CM, Green MJ. DNA methylation in peripheral tissue of schizophrenia and bipolar disorder: a systematic review. BMC Genet 2016; 17:27. [PMID: 26809779 PMCID: PMC4727379 DOI: 10.1186/s12863-016-0332-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/13/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increasing evidence suggests the involvement of epigenetic processes in the development of schizophrenia and bipolar disorder, and recent reviews have focused on findings in post-mortem brain tissue. A systematic review was conducted to synthesise and evaluate the quality of available evidence for epigenetic modifications (specifically DNA methylation) in peripheral blood and saliva samples of schizophrenia and bipolar disorder patients in comparison to healthy controls. METHODS Original research articles using humans were identified using electronic databases. There were 33 included studies for which data were extracted and graded in duplicate on 22 items of the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement, to assess methodological precision and quality of reporting. RESULTS There were 15 genome-wide and 18 exclusive candidate gene loci investigations for DNA methylation studies. A number of common genes were identified as differentially methylated in schizophrenia/bipolar disorder, which were related to reelin, brain-derived neurotrophic factor, dopamine (including the catechol-O-methyltransferase gene), serotonin and glutamate, despite inconsistent findings of hyper-, hypo-, or lack of methylation at these and other loci. The mean STROBE score of 59% suggested moderate quality of available evidence; however, wide methodological variability contributed to a lack of consistency in the way methylation levels were quantified, such that meta-analysis of the results was not possible. CONCLUSIONS Moderate quality of available evidence shows some convergence of differential methylation at some common genetic loci in schizophrenia and bipolar disorder, despite wide variation in methodology and reporting across studies. Improvement in the clarity of reporting clinical and other potential confounds would be useful in future studies of epigenetic processes in the context of exposure to environmental and other risk factors.
Collapse
Affiliation(s)
- Nina Teroganova
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia.
- Schizophrenia Research Institute, 405 Liverpool St, Darlinghurst, NSW, 2010, Australia.
| | - Leah Girshkin
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia.
- Schizophrenia Research Institute, 405 Liverpool St, Darlinghurst, NSW, 2010, Australia.
| | - Catherine M Suter
- Molecular Structural and Computational Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.
| | - Melissa J Green
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia.
- Schizophrenia Research Institute, 405 Liverpool St, Darlinghurst, NSW, 2010, Australia.
- Neuroscience Research Australia, Sydney, NSW, 2031, Australia.
| |
Collapse
|
32
|
Inoshita M, Numata S, Tajima A, Kinoshita M, Umehara H, Yamamori H, Hashimoto R, Imoto I, Ohmori T. Sex differences of leukocytes DNA methylation adjusted for estimated cellular proportions. Biol Sex Differ 2015; 6:11. [PMID: 26113971 PMCID: PMC4480901 DOI: 10.1186/s13293-015-0029-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 06/06/2015] [Indexed: 12/31/2022] Open
Abstract
Background DNA methylation, which is most frequently the transference of a methyl group to the 5-carbon position of the cytosine in a CpG dinucleotide, plays an important role in both normal development and diseases. To date, several genome-wide methylome studies have revealed sex-biased DNA methylation, yet no studies have investigated sex differences in DNA methylation by taking into account cellular heterogeneity. The aim of the present study was to investigate sex-biased DNA methylation on the autosomes in human blood by adjusting for estimated cellular proportions because cell-type proportions may vary by sex. Methods We performed a genome-wide DNA methylation profiling of the peripheral leukocytes in two sets of samples, a discovery set (49 males and 44 females) and a replication set (14 males and 10 females) using Infinium HumanMethylation450 BeadChips for 485,764 CpG dinucleotides and then examined the effect of sex on DNA methylation with a multiple linear regression analysis after adjusting for age, the estimated 6 cell-type proportions, and the covariates identified in a surrogate variable analysis. Results We identified differential DNA methylation between males and females at 292 autosomal CpG site loci in the discovery set (Bonferroni-adjusted p < 0.05). Of these 292 CpG sites, significant sex differences were also observed at 98 sites in the replication set (p < 0.05). Conclusions These findings provided further evidence that DNA methylation may play a role in the differentiation or maintenance of sexual dimorphisms. Our methylome mapping of the effects of sex may be useful to understanding the molecular mechanism involved in both normal development and diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13293-015-0029-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masatoshi Inoshita
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima, 770-8503 Japan
| | - Shusuke Numata
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima, 770-8503 Japan
| | - Atsushi Tajima
- Department of Human Genetics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima, 770-8503 Japan ; Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8640 Japan
| | - Makoto Kinoshita
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima, 770-8503 Japan
| | - Hidehiro Umehara
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima, 770-8503 Japan
| | - Hidenaga Yamamori
- Department of Molecular Neuropsychiatry, Osaka University Graduate School of Medicine, Suita, Osaka 5650871 Japan ; Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka 5650871 Japan
| | - Ryota Hashimoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka 5650871 Japan ; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka 5650871 Japan
| | - Issei Imoto
- Department of Human Genetics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima, 770-8503 Japan
| | - Tetsuro Ohmori
- Department of Psychiatry, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto, Tokushima, 770-8503 Japan
| |
Collapse
|
33
|
Abdolmaleky HM, Zhou JR, Thiagalingam S. An update on the epigenetics of psychotic diseases and autism. Epigenomics 2015; 7:427-49. [DOI: 10.2217/epi.14.85] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The examination of potential roles of epigenetic alterations in the pathogenesis of psychotic diseases have become an essential alternative in recent years as genetic studies alone are yet to uncover major gene(s) for psychosis. Here, we describe the current state of knowledge from the gene-specific and genome-wide studies of postmortem brain and blood cells indicating that aberrant DNA methylation, histone modifications and dysregulation of micro-RNAs are linked to the pathogenesis of mental diseases. There is also strong evidence supporting that all classes of psychiatric drugs modulate diverse features of the epigenome. While comprehensive environmental and genetic/epigenetic studies are uncovering the origins, and the key genes/pathways affected in psychotic diseases, characterizing the epigenetic effects of psychiatric drugs may help to design novel therapies in psychiatry.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Departments of Medicine (Biomedical Genetics Section), Genetics & Genomics, Boston University School of Medicine, Boston, MA 02118, USA
- Nutrition/Metabolism Laboratory at Beth Israel Deaconess Medical Center, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory at Beth Israel Deaconess Medical Center, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Sam Thiagalingam
- Departments of Medicine (Biomedical Genetics Section), Genetics & Genomics, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
34
|
Tseng PT, Lin PY, Lee Y, Hung CF, Lung FW, Chen CS, Chong MY. Age-associated decrease in global DNA methylation in patients with major depression. Neuropsychiatr Dis Treat 2014; 10:2105-14. [PMID: 25419133 PMCID: PMC4235206 DOI: 10.2147/ndt.s71997] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Evidence has supported a role of DNA methylation in the pathophysiology of mood disorders. The purpose of the current study is to examine 5-methylcytosine (5-mc) and 5-hydroxymethylcytosine (5-hmc) levels in patients with major depressive disorder (MDD) at different disease states. METHODS Forty-nine patients with MDD and 25 healthy control subjects were included. The severity in the disease was assessed by using the 17-item Hamilton Rating Scale of Depression (HAM-D) (HAM-D ≥19 for severe MDD and HAM-D ≤7 for remitted MDD). The 5-mc and 5-hmc levels in leukocyte DNA were measured using an enzyme-linked immunosorbent assay-based method. RESULTS We found a significant decrease in 5-hmc and trends of decreasing 5-mc levels in patients with severe MDD compared to healthy controls (P=0.059 for 5-mc and P=0.013 for 5-hmc). The decrease in the level exists only in the older age group (P=0.035 for 5-mc and P=0.002 for 5-hmc) but not in the younger age group (P=0.077 for 5-mc and P=0.620 for 5-hmc). In addition, the 5-mc level was found to be inversely correlated with disease severity (P=0.011). CONCLUSION Our results support a decrease in global DNA methylation associated with age in patients with severe depression. Further studies are needed to clarify the role of the methylation level as a disease marker of depression and whether antidepressant treatment changes the methylation profiles.
Collapse
Affiliation(s)
- Ping-Tao Tseng
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan ; Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung Jen-Ai's Home, Taiwan
| | - Pao-Yen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan ; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu Lee
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chi-Fa Hung
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - For-Wey Lung
- Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan ; Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan ; Department of Psychiatry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mian-Yoon Chong
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|