0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Genome-wide DNA methylation risk scores for schizophrenia derived from blood and brain tissues further explain the genetic risk in patients stratified by polygenic risk scores for schizophrenia and bipolar disorder

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Genetic and environmental factors contribute to the pathogenesis of schizophrenia (SZ) and bipolar disorder (BD). Among genetic risk groups stratified by combinations of Polygenic Risk Score (PRS) deciles for SZ, BD and SZ versus BD, genetic SZ risk groups had high SZ risk and prominent cognitive impairments. Furthermore, epigenetic alterations are implicated in these disorders. However, it was unclear whether DNA Methylation Risk Scores (MRSs) for SZ risk derived from blood and brain tissues were associated with SZ risk, particularly the PRS-stratified genetic SZ risk group.

          Methods

          Epigenome-wide association studies (EWASs) of SZ risk in whole blood were preliminarily conducted between 66 SZ patients and 30 healthy controls (HCs) and among genetic risk groups (individuals with low genetic risk for SZ and BD in HCs (n=30) and in SZ patients (n=11), genetic BD risk in SZ patients (n=25) and genetic SZ risk in SZ patients (n=30)) stratified by combinations of PRSs for SZ, BD and SZ versus BD. Next, differences in MRSs based on independent EWASs of SZ risk in whole blood, postmortem frontal cortex (FC) and superior temporal gyrus (STG) were investigated among our case‒control and PRS-stratified genetic risk status groups.

          Results

          Among case‒control and genetic risk status groups, 33 and 351 genome-wide significant differentially methylated positions (DMPs) associated with SZ were identified, respectively, many of which were hypermethylated. Compared with the low genetic risk in HCs group, the genetic SZ risk in SZ group had 39 genome-wide significant DMPs, while the genetic BD risk in SZ group had only six genome-wide significant DMPs. The MRSs for SZ risk derived from whole blood, FC and STG were higher in our SZ patients than in HCs in whole blood and were particularly higher in the genetic SZ risk in SZ group than in the low genetic risk in HCs and genetic BD risk in SZ groups. Conversely, the MRSs for SZ risk based on our whole-blood EWASs among genetic risk groups were also associated with SZ in the FC and STG. There were no correlations between the MRSs and PRSs.

          Conclusions

          These findings suggest that the MRS is a potential genetic marker in understanding SZ, particularly in patients with a genetic SZ risk.

          Related collections

          Most cited references57

          • Record: found
          • Abstract: found
          • Article: not found

          DNA methylation and its basic function.

          In the mammalian genome, DNA methylation is an epigenetic mechanism involving the transfer of a methyl group onto the C5 position of the cytosine to form 5-methylcytosine. DNA methylation regulates gene expression by recruiting proteins involved in gene repression or by inhibiting the binding of transcription factor(s) to DNA. During development, the pattern of DNA methylation in the genome changes as a result of a dynamic process involving both de novo DNA methylation and demethylation. As a consequence, differentiated cells develop a stable and unique DNA methylation pattern that regulates tissue-specific gene transcription. In this chapter, we will review the process of DNA methylation and demethylation in the nervous system. We will describe the DNA (de)methylation machinery and its association with other epigenetic mechanisms such as histone modifications and noncoding RNAs. Intriguingly, postmitotic neurons still express DNA methyltransferases and components involved in DNA demethylation. Moreover, neuronal activity can modulate their pattern of DNA methylation in response to physiological and environmental stimuli. The precise regulation of DNA methylation is essential for normal cognitive function. Indeed, when DNA methylation is altered as a result of developmental mutations or environmental risk factors, such as drug exposure and neural injury, mental impairment is a common side effect. The investigation into DNA methylation continues to show a rich and complex picture about epigenetic gene regulation in the central nervous system and provides possible therapeutic targets for the treatment of neuropsychiatric disorders.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Functions of DNA methylation: islands, start sites, gene bodies and beyond.

            DNA methylation is frequently described as a 'silencing' epigenetic mark, and indeed this function of 5-methylcytosine was originally proposed in the 1970s. Now, thanks to improved genome-scale mapping of methylation, we can evaluate DNA methylation in different genomic contexts: transcriptional start sites with or without CpG islands, in gene bodies, at regulatory elements and at repeat sequences. The emerging picture is that the function of DNA methylation seems to vary with context, and the relationship between DNA methylation and transcription is more nuanced than we realized at first. Improving our understanding of the functions of DNA methylation is necessary for interpreting changes in this mark that are observed in diseases such as cancer.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mapping genomic loci implicates genes and synaptic biology in schizophrenia

              Schizophrenia has a heritability of 60-80%1, much of which is attributable to common risk alleles. Here, in a two-stage genome-wide association study of up to 76,755 individuals with schizophrenia and 243,649 control individuals, we report common variant associations at 287 distinct genomic loci. Associations were concentrated in genes that are expressed in excitatory and inhibitory neurons of the central nervous system, but not in other tissues or cell types. Using fine-mapping and functional genomic data, we identify 120 genes (106 protein-coding) that are likely to underpin associations at some of these loci, including 16 genes with credible causal non-synonymous or untranslated region variation. We also implicate fundamental processes related to neuronal function, including synaptic organization, differentiation and transmission. Fine-mapped candidates were enriched for genes associated with rare disruptive coding variants in people with schizophrenia, including the glutamate receptor subunit GRIN2A and transcription factor SP4, and were also enriched for genes implicated by such variants in neurodevelopmental disorders. We identify biological processes relevant to schizophrenia pathophysiology; show convergence of common and rare variant associations in schizophrenia and neurodevelopmental disorders; and provide a resource of prioritized genes and variants to advance mechanistic studies.
                Bookmark

                Author and article information

                Journal
                BMJ Ment Health
                BMJ Ment Health
                bmjment
                ebmh
                BMJ Mental Health
                BMJ Publishing Group (BMA House, Tavistock Square, London, WC1H 9JR )
                2755-9734
                2024
                12 January 2024
                : 27
                : 1
                : e300936
                Affiliations
                [1 ] departmentDepartment of Psychiatry , Ringgold_38225Gifu University Graduate School of Medicine , Gifu, Japan
                [2 ] departmentDepartment of General Internal Medicine , Kanazawa Medical University , Ishikawa, Japan
                [3 ] departmentGenome Medical Science Project (Toyama) , Ringgold_13805National Center for Global Health and Medicine (NCGM) , Tokyo, Japan
                [4 ] departmentLaboratory of Pharmaceutics, Department of Biomedical Pharmaceutics , Ringgold_12784Gifu Pharmaceutical University , Gifu, Japan
                [5 ] departmentAddictive Substance Project , Ringgold_13931Tokyo Metropolitan Institute of Medical Science , Tokyo, Japan
                Author notes
                [Correspondence to ] Dr Kazutaka Ohi, Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan; ohi.kazutaka.h8@ 123456f.gifu-u.ac.jp
                Author information
                http://orcid.org/0000-0001-9577-9640
                Article
                bmjment-2023-300936
                10.1136/bmjment-2023-300936
                10806921
                38216218
                2a8e0077-6c08-49dc-a3c1-09e7c4f2e52b
                © Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. Published by BMJ.

                This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/.

                History
                : 09 November 2023
                : 14 December 2023
                Funding
                Funded by: Grants-in-Aid for Young Scientists (B) (16K19784) from the Japan Society for the Promotion of Science (JSPS);
                Award ID: 16K19784
                Funded by: Grants-in-Aid for Scientific Research (C) from the Japan Society for the Promotion of Science (JSPS);
                Award ID: (19K08081, 21K07497, and 22K07614)
                Funded by: SENSHIN Medical Research Foundation;
                Award ID: N/A
                Funded by: AMED;
                Award ID: JP21uk1024002, JP22dk0307112 and JP23dk0307103
                Funded by: YOKOYAMA Foundation for Clinical Pharmacology;
                Award ID: YRY-1807
                Funded by: FundRef http://dx.doi.org/10.13039/100008732, Uehara Memorial Foundation;
                Award ID: N/A
                Funded by: FundRef http://dx.doi.org/10.13039/501100005865, Mochida Memorial Foundation for Medical and Pharmaceutical Research;
                Award ID: N/A
                Funded by: FundRef http://dx.doi.org/10.13039/501100004330, Smoking Research Foundation;
                Award ID: N/A
                Funded by: FundRef http://dx.doi.org/10.13039/100007449, Takeda Science Foundation;
                Award ID: N/A
                Categories
                Genetics and Genomics
                1506
                Custom metadata
                unlocked

                psychiatry,schizophrenia & psychotic disorders,depression & mood disorders

                Comments

                Comment on this article