5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Homocysteine facilitates endoplasmic reticulum stress and apoptosis of hepatocytes by suppressing ERO1α expression via cooperation between DNMT1 and G9a

      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

          Endoplasmic reticulum (ER) stress and apoptosis play a critical role in liver injury. Endoplasmic reticulum oxidoreductase 1α (ERO1α) is an oxidase that exists in the luminal side of the ER membrane, participating in protein folding and secretion and inhibiting apoptosis, but the underlying mechanism on liver injury induced by homocysteine (Hcy) remains obscure. In this study, hyperhomocysteinemia (HHcy) mice model was established in cbs +/− mice by feeding a high‐methionine diet for 12 weeks; and cbs +/− mice fed with high‐methionine diet exhibited more severe liver injury compared to cbs +/+ mice. Mechanistically, we found that Hcy promoted ER stress and apoptosis of hepatocytes and thereby aggravated liver injury through inhibiting ERO1α expression; accordingly, overexpression of ERO1α remarkably alleviated ER stress and apoptosis of hepatocytes induced by Hcy. Epigenetic modification analysis revealed that Hcy significantly increased levels of DNA methylation and H3 lysine 9 dimethylation (H3K9me2) on ERO1α promoter, which attributed to upregulated DNA methyltransferase 1 (DNMT1) and G9a, respectively. Further study showed that DNMT1 and G9a cooperatively regulated ERO1α expression in hepatocytes exposed to Hcy. Taken together, our work demonstrates that Hcy activates ER stress and apoptosis of hepatocytes by downregulating ERO1α expression via cooperation between DNMT1 and G9a, which provides new insight into the mechanism of Hcy‐induced ER stress and apoptosis of hepatocytes in liver injury.

          Related collections

          Most cited references34

          • 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

            Endoplasmic reticulum stress in liver disease.

            The unfolded protein response (UPR) is activated upon the accumulation of misfolded proteins in the endoplasmic reticulum (ER) that are sensed by the binding immunoglobulin protein (BiP)/glucose-regulated protein 78 (GRP78). The accumulation of unfolded proteins sequesters BiP so it dissociates from three ER-transmembrane transducers leading to their activation. These transducers are inositol requiring (IRE) 1α, PKR-like ER kinase (PERK), and activating transcription factor (ATF) 6α. PERK phosphorylates eukaryotic initiation factor 2 alpha (eIF2α) resulting in global mRNA translation attenuation, and concurrently selectively increases the translation of several mRNAs, including the transcription factor ATF4, and its downstream target CHOP. IRE1α has kinase and endoribonuclease (RNase) activities. IRE1α autophosphorylation activates the RNase activity to splice XBP1 mRNA, to produce the active transcription factor sXBP1. IRE1α activation also recruits and activates the stress kinase JNK. ATF6α transits to the Golgi compartment where it is cleaved by intramembrane proteolysis to generate a soluble active transcription factor. These UPR pathways act in concert to increase ER content, expand the ER protein folding capacity, degrade misfolded proteins, and reduce the load of new proteins entering the ER. All of these are geared toward adaptation to resolve the protein folding defect. Faced with persistent ER stress, adaptation starts to fail and apoptosis occurs, possibly mediated through calcium perturbations, reactive oxygen species, and the proapoptotic transcription factor CHOP. The UPR is activated in several liver diseases; including obesity associated fatty liver disease, viral hepatitis, and alcohol-induced liver injury, all of which are associated with steatosis, raising the possibility that ER stress-dependent alteration in lipid homeostasis is the mechanism that underlies the steatosis. Hepatocyte apoptosis is a pathogenic event in several liver diseases, and may be linked to unresolved ER stress. If this is true, restoration of ER homeostasis prior to ER stress-induced cell death may provide a therapeutic rationale in these diseases. Herein we discuss each branch of the UPR and how they may impact hepatocyte function in different pathologic states. Copyright © 2010 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              DNA hypomethylation in cancer cells.

              DNA hypomethylation was the initial epigenetic abnormality recognized in human tumors. However, for several decades after its independent discovery by two laboratories in 1983, it was often ignored as an unwelcome complication, with almost all of the attention on the hypermethylation of promoters of genes that are silenced in cancers (e.g., tumor-suppressor genes). Because it was subsequently shown that global hypomethylation of DNA in cancer was most closely associated with repeated DNA elements, cancer linked-DNA hypomethylation continued to receive rather little attention. DNA hypomethylation in cancer can no longer be considered an oddity, because recent high-resolution genome-wide studies confirm that DNA hypomethylation is the almost constant companion to hypermethylation of the genome in cancer, just usually (but not always) in different sequences. Methylation changes at individual CpG dyads in cancer can have a high degree of dependence not only on the regional context, but also on neighboring sites. DNA demethylation during carcinogenesis may involve hemimethylated dyads as intermediates, followed by spreading of the loss of methylation on both strands. In this review, active demethylation of DNA and the relationship of cancer-associated DNA hypomethylation to cancer stem cells are discussed. Evidence is accumulating for the biological significance and clinical relevance of DNA hypomethylation in cancer, and for cancer-linked demethylation and de novo methylation being highly dynamic processes.
                Bookmark

                Author and article information

                Contributors
                zhp19760820@163.com
                jydeng@nxmu.edu.cn
                Journal
                Cell Biol Int
                Cell Biol Int
                10.1002/(ISSN)1095-8355
                CBIN
                Cell Biology International
                John Wiley and Sons Inc. (Hoboken )
                1065-6995
                1095-8355
                14 April 2022
                August 2022
                : 46
                : 8 ( doiID: 10.1002/cbin.v46.8 )
                : 1236-1248
                Affiliations
                [ 1 ] NHC Key Laboratory of Metabolic Cardiovascular Diseases Research Ningxia Medical University Yinchuan China
                [ 2 ] Ningxia Key Laboratory of Vascular Injury and Repair Research Ningxia Medical University Yinchuan China
                [ 3 ] Department of Clinical Medicine General Hospital of Ningxia Medical University Yinchuan China
                [ 4 ] Department of Infectious Diseases General Hospital of Ningxia Medical University Yinchuan China
                [ 5 ] School of Basic Medical Sciences Ningxia Medical University Yinchuan China
                [ 6 ] Department of Prenatal Diagnosis Center General Hospital of Ningxia Medical University Yinchuan China
                Author notes
                [*] [* ] Correspondence: Huiping Zhang, Prenatal Diagnosis Center, General Hospital of Ningxia Medical University, Shengli St 804, Yinchuan 750004, China.

                Email: zhp19760820@ 123456163.com

                Yideng Jiang, School of Basic Medical Sciences, Ningxia Medical University, Shengli St 1160, Yinchuan 750004, China.

                Email: jydeng@ 123456nxmu.edu.cn

                Author information
                http://orcid.org/0000-0002-4882-5197
                Article
                CBIN11805
                10.1002/cbin.11805
                9543485
                35347798
                632a1ce7-c51d-418c-b414-431597e69953
                © 2022 The Authors. Cell Biology International published by John Wiley & Sons Ltd on behalf of International Federation of Cell Biology.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 16 March 2022
                : 04 November 2021
                : 27 March 2022
                Page count
                Figures: 5, Tables: 2, Pages: 13, Words: 7168
                Funding
                Funded by: Key Research and Development Projects in Ningxia Province
                Award ID: 2018BEG02004
                Award ID: 2019BFG02004
                Award ID: 2020BEG03005
                Award ID: 2020BFH02003
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: 81870225
                Award ID: 81760139
                Award ID: 81860555
                Award ID: 81870332
                Award ID: 82060110
                Funded by: Natural Science Foundation of Ningxia Province , doi 10.13039/501100004772;
                Award ID: 2020AAC02021
                Award ID: 2020AAC02038
                Award ID: 2021AAC03337
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                August 2022
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.2.0 mode:remove_FC converted:07.10.2022

                Cell biology
                apoptosis,dnmt1,endoplasmic reticulum stress,ero1α,g9a,homocysteine
                Cell biology
                apoptosis, dnmt1, endoplasmic reticulum stress, ero1α, g9a, homocysteine

                Comments

                Comment on this article