1
|
Li S, Mehal WZ, Ouyang X. RNA modifications in the progression of liver diseases: from fatty liver to cancer. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2105-2119. [PMID: 38809498 DOI: 10.1007/s11427-023-2494-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/24/2023] [Indexed: 05/30/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a prominent global health concern associated with high risk of metabolic syndrome, and has impacted a substantial segment of the population. The disease spectrum ranges from simple fatty liver to non-alcoholic steatohepatitis (NASH), which can progress to cirrhosis and hepatocellular carcinoma (HCC) and is increasingly becoming a prevalent indication for liver transplantation. The existing therapeutic options for NAFLD, NASH, and HCC are limited, underscoring the urgent need for innovative treatment strategies. Insights into gene expression, particularly RNA modifications such as N6 methyladenosine (m6A), hold promising avenues for interventions. These modifications play integral roles in RNA metabolism and cellular functions, encompassing the entire NAFLD-NASH-HCC progression. This review will encompass recent insights on diverse RNA modifications, including m6A, pseudouridine (ψ), N1-methyladenosine (m1A), and 5-methylcytidine (m5C) across various RNA species. It will uncover their significance in crucial aspects such as steatosis, inflammation, fibrosis, and tumorigenesis. Furthermore, prospective research directions and therapeutic implications will be explored, advancing our comprehensive understanding of the intricate interconnected nature of these pathological conditions.
Collapse
Affiliation(s)
- Simiao Li
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Wajahat Z Mehal
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Xinshou Ouyang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
2
|
Zha X, Gao Z, Li M, Xia X, Mao Z, Wang S. Insight into the regulatory mechanism of m 6A modification: From MAFLD to hepatocellular carcinoma. Biomed Pharmacother 2024; 177:116966. [PMID: 38906018 DOI: 10.1016/j.biopha.2024.116966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
In recent years, there has been a significant increase in the incidence of metabolic-associated fatty liver disease (MAFLD), which has been attributed to the increasing prevalence of type 2 diabetes mellitus (T2DM) and obesity. MAFLD affects more than one-third of adults worldwide, making it the most prevalent liver disease globally. Moreover, MAFLD is considered a significant risk factor for hepatocellular carcinoma (HCC), with MAFLD-related HCC cases increasing. Approximately 1 in 6 HCC patients are believed to have MAFLD, and nearly 40 % of these HCC patients do not progress to cirrhosis, indicating direct transformation from MAFLD to HCC. N6-methyladenosine (m6A) is commonly distributed in eukaryotic mRNA and plays a crucial role in normal development and disease progression, particularly in tumors. Numerous studies have highlighted the close association between abnormal m6A modification and cellular metabolic alterations, underscoring its importance in the onset and progression of MAFLD. However, the specific impact of m6A modification on the progression of MAFLD to HCC remains unclear. Can targeting m6A effectively halt the progression of MAFLD-related HCC? In this review, we investigated the pivotal role of abnormal m6A modification in the transition from MAFLD to HCC, explored the potential of m6A modification as a therapeutic target for MAFLD-related HCC, and proposed possible directions for future investigations.
Collapse
Affiliation(s)
- Xuan Zha
- Department of Laboratory Medicine, the Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zewei Gao
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Min Li
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xueli Xia
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhenwei Mao
- Department of Laboratory Medicine, Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
3
|
Melnik BC, Weiskirchen R, Stremmel W, John SM, Schmitz G. Risk of Fat Mass- and Obesity-Associated Gene-Dependent Obesogenic Programming by Formula Feeding Compared to Breastfeeding. Nutrients 2024; 16:2451. [PMID: 39125332 PMCID: PMC11314333 DOI: 10.3390/nu16152451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
It is the purpose of this review to compare differences in postnatal epigenetic programming at the level of DNA and RNA methylation and later obesity risk between infants receiving artificial formula feeding (FF) in contrast to natural breastfeeding (BF). FF bears the risk of aberrant epigenetic programming at the level of DNA methylation and enhances the expression of the RNA demethylase fat mass- and obesity-associated gene (FTO), pointing to further deviations in the RNA methylome. Based on a literature search through Web of Science, Google Scholar, and PubMed databases concerning the dietary and epigenetic factors influencing FTO gene and FTO protein expression and FTO activity, FTO's impact on postnatal adipogenic programming was investigated. Accumulated translational evidence underscores that total protein intake as well as tryptophan, kynurenine, branched-chain amino acids, milk exosomal miRNAs, NADP, and NADPH are crucial regulators modifying FTO gene expression and FTO activity. Increased FTO-mTORC1-S6K1 signaling may epigenetically suppress the WNT/β-catenin pathway, enhancing adipocyte precursor cell proliferation and adipogenesis. Formula-induced FTO-dependent alterations of the N6-methyladenosine (m6A) RNA methylome may represent novel unfavorable molecular events in the postnatal development of adipogenesis and obesity, necessitating further investigations. BF provides physiological epigenetic DNA and RNA regulation, a compelling reason to rely on BF.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Wolfgang Stremmel
- Praxis for Internal Medicine, Beethovenstrasse 2, D-76530 Baden-Baden, Germany;
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Institute for Interdisciplinary Dermatological Prevention and Rehabilitation (iDerm), University of Osnabrück, D-49076 Osnabrück, Germany;
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
4
|
Han X, Zhu Y, Ke J, Zhai Y, Huang M, Zhang X, He H, Zhang X, Zhao X, Guo K, Li X, Han Z, Zhang Y. Progression of m 6A in the tumor microenvironment: hypoxia, immune and metabolic reprogramming. Cell Death Discov 2024; 10:331. [PMID: 39033180 PMCID: PMC11271487 DOI: 10.1038/s41420-024-02092-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
Recently, N6-methyladenosine (m6A) has aroused widespread discussion in the scientific community as a mode of RNA modification. m6A comprises writers, erasers, and readers, which regulates RNA production, nuclear export, and translation and is very important for human health. A large number of studies have found that the regulation of m6A is closely related to the occurrence and invasion of tumors, while the homeostasis and function of the tumor microenvironment (TME) determine the occurrence and development of tumors to some extent. TME is composed of a variety of immune cells (T cells, B cells, etc.) and nonimmune cells (tumor-associated mesenchymal stem cells (TA-MSCs), cancer-associated fibroblasts (CAFs), etc.). Current studies suggest that m6A is involved in regulating the function of various cells in the TME, thereby affecting tumor progression. In this manuscript, we present the composition of m6A and TME, the relationship between m6A methylation and characteristic changes in TME, the role of m6A methylation in TME, and potential therapeutic strategies to provide new perspectives for better treatment of tumors in clinical work.
Collapse
Affiliation(s)
- Xuan Han
- First Clinical College of Changzhi Medical College, Changzhi, China
| | - Yu Zhu
- Linfen Central Hospital, Linfen, China
| | - Juan Ke
- Linfen Central Hospital, Linfen, China
| | | | - Min Huang
- Linfen Central Hospital, Linfen, China
| | - Xin Zhang
- Linfen Central Hospital, Linfen, China
| | | | | | | | | | | | - Zhongyu Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | |
Collapse
|
5
|
Li A, Wang R, Zhao Y, Zhao P, Yang J. Crosstalk between Epigenetics and Metabolic Reprogramming in Metabolic Dysfunction-Associated Steatotic Liver Disease-Induced Hepatocellular Carcinoma: A New Sight. Metabolites 2024; 14:325. [PMID: 38921460 PMCID: PMC11205353 DOI: 10.3390/metabo14060325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Epigenetic and metabolic reprogramming alterations are two important features of tumors, and their reversible, spatial, and temporal regulation is a distinctive hallmark of carcinogenesis. Epigenetics, which focuses on gene regulatory mechanisms beyond the DNA sequence, is a new entry point for tumor therapy. Moreover, metabolic reprogramming drives hepatocellular carcinoma (HCC) initiation and progression, highlighting the significance of metabolism in this disease. Exploring the inter-regulatory relationship between tumor metabolic reprogramming and epigenetic modification has become one of the hot directions in current tumor metabolism research. As viral etiologies have given way to metabolic dysfunction-associated steatotic liver disease (MASLD)-induced HCC, it is urgent that complex molecular pathways linking them and hepatocarcinogenesis be explored. However, how aberrant crosstalk between epigenetic modifications and metabolic reprogramming affects MASLD-induced HCC lacks comprehensive understanding. A better understanding of their linkages is necessary and urgent to improve HCC treatment strategies. For this reason, this review examines the interwoven landscape of molecular carcinogenesis in the context of MASLD-induced HCC, focusing on mechanisms regulating aberrant epigenetic alterations and metabolic reprogramming in the development of MASLD-induced HCC and interactions between them while also updating the current advances in metabolism and epigenetic modification-based therapeutic drugs in HCC.
Collapse
Affiliation(s)
- Anqi Li
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (A.L.); (Y.Z.); (P.Z.)
| | - Rui Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Yuqiang Zhao
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (A.L.); (Y.Z.); (P.Z.)
| | - Peiran Zhao
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (A.L.); (Y.Z.); (P.Z.)
| | - Jing Yang
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (A.L.); (Y.Z.); (P.Z.)
| |
Collapse
|
6
|
Chen A, Zhang VX, Zhang Q, Sze KMF, Tian L, Huang H, Wang X, Lee E, Lu J, Lyu X, Lee MFJ, Wong CM, Ho DWH, Ng IOL. Targeting the oncogenic m6A demethylase FTO suppresses tumourigenesis and potentiates immune response in hepatocellular carcinoma. Gut 2024:gutjnl-2024-331903. [PMID: 38839271 DOI: 10.1136/gutjnl-2024-331903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024]
Abstract
OBJECTIVE Fat mass and obesity-associated protein (FTO), an eraser of N 6-methyadenosine (m6A), plays oncogenic roles in various cancers. However, its role in hepatocellular carcinoma (HCC) is unclear. Furthermore, small extracellular vesicles (sEVs, or exosomes) are critical mediators of tumourigenesis and metastasis, but the relationship between FTO-mediated m6A modification and sEVs in HCC is unknown. DESIGN The functions and mechanisms of FTO and glycoprotein non-metastatic melanoma protein B (GPNMB) in HCC progression were investigated in vitro and in vivo. Neutralising antibody of syndecan-4 (SDC4) was used to assess the significance of sEV-GPNMB. FTO inhibitor CS2 was used to examine the effects on anti-PD-1 and sorafenib treatment. RESULTS FTO expression was upregulated in patient HCC tumours. Functionally, FTO promoted HCC cell proliferation, migration and invasion in vitro, and tumour growth and metastasis in vivo. FTO knockdown enhanced the activation and recruitment of tumour-infiltrating CD8+ T cells. Furthermore, we identified GPNMB to be a downstream target of FTO, which reduced the m6A abundance of GPNMB, hence, stabilising it from degradation by YTH N 6-methyladenosine RNA binding protein F2. Of note, GPNMB was packaged into sEVs derived from HCC cells and bound to the surface receptor SDC4 of CD8+ T cells, resulting in the inhibition of CD8+ T cell activation. A potential FTO inhibitor, CS2, suppresses the oncogenic functions of HCC cells and enhances the sensitivity of anti-PD-1 and sorafenib treatment. CONCLUSION Targeting the FTO/m6A/GPNMB axis could significantly suppress tumour growth and metastasis, and enhance immune activation, highlighting the potential of targeting FTO signalling with effective inhibitors for HCC therapy.
Collapse
Affiliation(s)
- Ao Chen
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
- Department of Biology, Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Vanilla Xin Zhang
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Qingyang Zhang
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Karen Man-Fong Sze
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Lu Tian
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Hongyang Huang
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Xia Wang
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Eva Lee
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jingyi Lu
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Xueying Lyu
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Man-Fong Joyce Lee
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Chun Ming Wong
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Daniel Wai-Hung Ho
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
7
|
Ming X, Chen S, Li H, Wang Y, Zhou L, Lv Y. m6A RNA Methylation and Implications for Hepatic Lipid Metabolism. DNA Cell Biol 2024; 43:271-278. [PMID: 38635960 DOI: 10.1089/dna.2023.0410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
This review presents a summary of recent progress in research on the N6-methyladenosine (m6A) modification and regulatory roles in hepatic lipid metabolism. As the most abundant internal modification of eukaryotic RNA, the m6A modification is a dynamic and reversible process of the m6A enzyme system, which includes writers, erasers, and readers. m6A methylation depressed lipid synthesis and facilitated lipolysis in liver. The depletion of m6A methyltransferase Mettl14/Mettl3 raised fatty acid synthase (FAS), stearoyl-CoA desaturase-1 (SCD1), acetyl-CoA carboxylase (ACC), and elongase of very long chain fatty acids 6 (ELOVL6) in rodent liver, causing increases in liver weight, triglyceride (TG) production, and content in hepatocytes. FTO catalyzed m6A demethylation and the suppression m6A reader YTHDC2 promoted hepatocellular TG generation and hepatic steatosis in C57BL/6 mice through sterol regulatory element-binding protein 1c (SREBP-1c) signaling pathway, which upregulated the lipogenic genes FAS, SCD1, ACC, recombinant acetyl coenzyme a carboxylase alpha, and cell death-inducing DNA fragmentation factor-like effector C (CIDEC). Furthermore, FTO overexpression did not only enhance mitochondrial fusion to impair mitochondrial function and lipid oxidation but also promoted lipid peroxidation, accompanied by excessive TG in hepatocytes and rodent liver. Elevated m6A modification potently suppressed hepatic lipid accumulation, while the shrinkage of m6A modification arose hepatic lipid deposition. These findings have highlighted the beneficial role of m6A RNA methylation in hepatic lipid metabolism, potentially protecting liver from lipid metabolic disorders.
Collapse
Affiliation(s)
- Xinyue Ming
- Guangxi Key Laboratory of Diabetic Systems Medicine, Faculty of Basic Medical Sciences, Institute of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Shirui Chen
- Guangxi Key Laboratory of Diabetic Systems Medicine, Faculty of Basic Medical Sciences, Institute of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Huijuan Li
- Guangxi Key Laboratory of Diabetic Systems Medicine, Faculty of Basic Medical Sciences, Institute of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Yun Wang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Faculty of Basic Medical Sciences, Institute of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Faculty of Basic Medical Sciences, Institute of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Yuncheng Lv
- Guangxi Key Laboratory of Diabetic Systems Medicine, Faculty of Basic Medical Sciences, Institute of Basic Medical Sciences, Guilin Medical University, Guilin, China
| |
Collapse
|
8
|
Chen SS, Zhang H. Abrogation and homeostatic restoration of IgE responses by a universal IgE allergy CTL vaccine-The three signal self/non-self/self (S/NS/S) theory. Immunology 2024; 172:91-108. [PMID: 38303079 PMCID: PMC10987285 DOI: 10.1111/imm.13753] [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: 04/26/2023] [Accepted: 12/06/2023] [Indexed: 02/03/2024] Open
Abstract
Natural IgE cytotoxic peptides (nECPs), which are derived from the constant domain of the heavy chain of human IgE producing B cells via endoplasmic reticulum (ER) stress, are decorated onto MHC class 1a molecules (MHCIa) as unique biomarkers for CTL (cytotoxic T lymphocyte)-mediated immune surveillance. Human IgE exhibits only one isotype and lacks polymorphisms; IgE is pivotal in mediating diverse, allergen-specific allergies. Therefore, by disrupting self-IgE tolerance via costimulation, the CTLs induced by nECPs can serve as universal allergy vaccines (UAVs) in humans to dampen IgE production mediated by diverse allergen-specific IgE-secreting B cells and plasma cells expressing surface nECP-MHCIa as targets. The study herein has enabled the identification of nECPs, A32 and SP-1/SP-2 nonameric natural peptides produced through the correspondence principle. Vaccination using nECP induced nECP-specific CTL that profoundly suppressed human IgE production in vitro as well as chimeric human IgE production in human IgE/HLA-A2.01/HLA-B7.02 triple transgenic rodents. Furthermore, nECP-tetramer-specific CTLs were found to be converted into CD4 Tregs that restored IgE competence via the homeostatic principle, mediatepred by SREBP-1c suppressed DCs. Thus, nECPs showed causal efficacy and safety as UAVs for treating categorically type I hypersensitivity IgE-mediated allergies. The applied vaccination concept presented provides the foundation to unify, integrate through a singular class of tetramer-specific TCR clonotypes for regulaing human IgE production. The three signal theory pertains to mechanisms of three cells underlying central tolerance (S), breaking self tolerance (NS) and regaining peripheral tolerance (S) via homeostasis concerning nECP as an efficacious and safe UAV to treat type I IgE-mediated hypersensitivity. The three signal theory impirically extended, may be heuritic for immuno-regulation of adaptive immune repertoire in general.
Collapse
Affiliation(s)
- Swey-Shen Chen
- Department of Immunology and Inflammation, AAIIT LLC, San Diego, California, USA
- Division of Vaccinology and Immunotherapy, IGE Therapeutics and Pharmaceuticals, Inc, San Diego, California, USA
- Department of Protein Display and Molecular Evolution, The Institute of Genetics at San Diego, San Diego, California, USA
| | - Hailan Zhang
- Department of Immunology and Inflammation, AAIIT LLC, San Diego, California, USA
- Division of Vaccinology and Immunotherapy, IGE Therapeutics and Pharmaceuticals, Inc, San Diego, California, USA
| |
Collapse
|
9
|
Guo J, Zhao L, Duan M, Yang Z, Zhao H, Liu B, Wang Y, Deng L, Wang C, Jiang X, Jiang X. Demethylases in tumors and the tumor microenvironment: Key modifiers of N 6-methyladenosine methylation. Biomed Pharmacother 2024; 174:116479. [PMID: 38537580 DOI: 10.1016/j.biopha.2024.116479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
RNA methylation modifications are widespread in eukaryotes and prokaryotes, with N6-methyladenosine (m6A) the most common among them. Demethylases, including Fat mass and obesity associated gene (FTO) and AlkB homolog 5 (ALKBH5), are important in maintaining the balance between RNA methylation and demethylation. Recent studies have clearly shown that demethylases affect the biological functions of tumors by regulating their m6A levels. However, their effects are complicated, and even opposite results have appeared in different articles. Here, we summarize the complex regulatory networks of demethylases, including the most important and common pathways, to clarify the role of demethylases in tumors. In addition, we describe the relationships between demethylases and the tumor microenvironment, and introduce their regulatory mechanisms. Finally, we discuss evaluation of demethylases for tumor diagnosis and prognosis, as well as the clinical application of demethylase inhibitors, providing a strong basis for their large-scale clinical application in the future.
Collapse
Affiliation(s)
- Junchen Guo
- Departmentof Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Liang Zhao
- Department of Anorectal Surgery, Shenyang Anorectal Hospital, Shenyang, Liaoning 110002, China
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - He Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Baiming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Yihan Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Liping Deng
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Chen Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Xiaodi Jiang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110002, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China.
| |
Collapse
|
10
|
Zhao L, Guo J, Xu S, Duan M, Liu B, Zhao H, Wang Y, Liu H, Yang Z, Yuan H, Jiang X, Jiang X. Abnormal changes in metabolites caused by m 6A methylation modification: The leading factors that induce the formation of immunosuppressive tumor microenvironment and their promising potential for clinical application. J Adv Res 2024:S2090-1232(24)00159-0. [PMID: 38677545 DOI: 10.1016/j.jare.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/14/2024] [Accepted: 04/14/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) RNA methylation modifications have been widely implicated in the metabolic reprogramming of various cell types within the tumor microenvironment (TME) and are essential for meeting the demands of cellular growth and maintaining tissue homeostasis, enabling cells to adapt to the specific conditions of the TME. An increasing number of research studies have focused on the role of m6A modifications in glucose, amino acid and lipid metabolism, revealing their capacity to induce aberrant changes in metabolite levels. These changes may in turn trigger oncogenic signaling pathways, leading to substantial alterations within the TME. Notably, certain metabolites, including lactate, succinate, fumarate, 2-hydroxyglutarate (2-HG), glutamate, glutamine, methionine, S-adenosylmethionine, fatty acids and cholesterol, exhibit pronounced deviations from normal levels. These deviations not only foster tumorigenesis, proliferation and angiogenesis but also give rise to an immunosuppressive TME, thereby facilitating immune evasion by the tumor. AIM OF REVIEW The primary objective of this review is to comprehensively discuss the regulatory role of m6A modifications in the aforementioned metabolites and their potential impact on the development of an immunosuppressive TME through metabolic alterations. KEY SCIENTIFIC CONCEPTS OF REVIEW This review aims to elaborate on the intricate networks governed by the m6A-metabolite-TME axis and underscores its pivotal role in tumor progression. Furthermore, we delve into the potential implications of the m6A-metabolite-TME axis for the development of novel and targeted therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- Liang Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Department of Colorectal Anal Surgery, Shenyang Coloproctology Hospital, Shenyang 110002, China.
| | - Junchen Guo
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Shasha Xu
- Department of Gastroendoscopy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Baiming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - He Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yihan Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Haiyang Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Hexue Yuan
- Department of Colorectal Anal Surgery, Shenyang Coloproctology Hospital, Shenyang 110002, China.
| | - Xiaodi Jiang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110020, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
11
|
Li J, Fang L, Xi M, Ni A, Qian Q, Wang Z, Wang H, Yan J. Toxic effects of triclosan on hepatic and intestinal lipid accumulation in zebrafish via regulation of m6A-RNA methylation. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 269:106884. [PMID: 38458066 DOI: 10.1016/j.aquatox.2024.106884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/21/2024] [Accepted: 03/01/2024] [Indexed: 03/10/2024]
Abstract
Triclosan (TCS), recognized as an endocrine disruptor, has raised significant concerns due to its widespread use and potential health risks. To explore the impact of TCS on lipid metabolism, both larval and adult zebrafish were subjected to acute and chronic exposure to TCS. Through analyzes of biochemical and physiological markers, as well as Oil Red O (ORO) and hematoxylin and eosin (H&E) staining, our investigation revealed that TCS exposure induced hepatic and intestinal lipid accumulation in larval and adult zebrafish, leading to structural damage and inflammatory responses in these tissues. The strong affinity of TCS with PPARγ and subsequent pathway activation indicate that PPARγ pathway plays a crucial role in TCS-induced lipid buildup. Furthermore, we observed a decrease in m6A-RNA methylation levels in the TCS-treated group, which attributed to the increased activity of the demethylase FTO and concurrent suppression of the methyltransferase METTL3 gene expression by TCS. The alteration in methylation dynamics is identified as a potential underlying mechanism behind TCS-induced lipid accumulation. To address this concern, we explored the impact of folic acid-a methyl donor for m6A-RNA methylation-on lipid accumulation in zebrafish. Remarkably, folic acid administration partially alleviated lipid accumulation by restoring m6A-RNA methylation. This restoration, in turn, contributed to a reduction in inflammatory damage observed in both the liver and intestines. Additionally, folic acid partially mitigates the up-regulation of PPARγ and related genes induced by TCS. These findings carry substantial implications for understanding the adverse effects of environmental pollutants such as TCS. They also emphasize the promising potential of folic acid as a therapeutic intervention to alleviate disturbances in lipid metabolism induced by environmental pollutants.
Collapse
Affiliation(s)
- Jinyun Li
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Lu Fang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Miaocui Xi
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Anyu Ni
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Qiuhui Qian
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Zejun Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Huili Wang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Jin Yan
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
12
|
Zhang Y, Chen L, Zhu J, Liu H, Xu L, Wu Y, He C, Song Y. Minor alleles of FTO rs9939609 and rs17817449 polymorphisms confer a higher risk of type 2 diabetes mellitus and dyslipidemia, but not coronary artery disease in a Chinese Han population. Front Endocrinol (Lausanne) 2023; 14:1249070. [PMID: 38161971 PMCID: PMC10754952 DOI: 10.3389/fendo.2023.1249070] [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: 08/02/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024] Open
Abstract
Background Relationships of the polymorphisms in fat mass and obesity-associated gene (FTO) and peroxisome proliferator-activated receptor delta gene (PPARD) with metabolic-related diseases remain to be clarified. Methods One thousand three hundred and eighty-one subjects were enrolled. Metabolic-related diseases including obesity, dyslipidemia, hyperhomocysteinemia, hyperuricemia, hypertension, type 2 diabetes mellitus (T2DM) and coronary artery disease (CAD) were defined based on diagnostic criteria. FTO rs9939609 and rs17817449, and PPARD rs2016520 and rs2267668 polymorphisms were genotyped by using polymerase chain reaction-restricted fragment length polymorphism method. Results Patients with T2DM or dyslipidemia had a higher frequency of AA, AT or AA + AT genotypes as well as A allele of FTO rs9939609 polymorphism than those free of T2DM or dyslipidemia (P ≤ 0.04 for all). Patients with T2DM or dyslipidemia had a higher frequency of GG, GT or GG + GT genotypes as well as G allele of FTO rs17817449 polymorphism than those free of T2DM or dyslipidemia (P ≤ 0.03 for all). Multivariate logistic regression analyses showed that FTO rs9939609 and rs17817449 polymorphisms were independently associated with T2DM as well as dyslipidemia after adjustment for age, sex, smoking and other metabolic diseases. FTO rs9939609 and rs17817449 polymorphisms were not associated with obesity, hyperhomocysteinemia, hyperuricemia, hypertension and CAD. Obese or T2DM carriers of the AA or AT genotype of the FTO rs9939609 polymorphism had a higher prevalence of dyslipidemia compared to non-obese or non-T2DM carriers of the AA or AT genotype (P = 0.03 for both). Among the carriers of GG or GT genotype of the FTO rs17817449 polymorphism, the prevalence of dyslipidemia in obese patients was higher than that in non-obese subjects (P < 0.01). PPARD rs2016520 and rs2267668 polymorphisms were not correlated with any of the metabolic-related diseases in the study population. Conclusion Minor alleles of FTO rs9939609 and rs17817449 polymorphisms confer a higher risk of T2DM and dyslipidemia, and the risk is further increased among obese individuals. PPARD rs2016520 and rs2267668 polymorphisms are not associated with metabolic-related diseases.
Collapse
Affiliation(s)
- Youjin Zhang
- Central Laboratory, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Lvlin Chen
- Department of Critical Care Medicine, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Junchen Zhu
- Department of Critical Care Medicine, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Hao Liu
- Clinical Medical College of Chengdu University, Chengdu, Sichuan, China
| | - Luying Xu
- Clinical Medical College of Chengdu University, Chengdu, Sichuan, China
| | - Yang Wu
- Clinical Medical College of Chengdu University, Chengdu, Sichuan, China
| | - Chuan He
- Department of Cardiology, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Yongyan Song
- Central Laboratory, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Luo P, Li S, Jing W, Tu J, Long X. N 6-methyladenosine RNA modification in nonalcoholic fatty liver disease. Trends Endocrinol Metab 2023; 34:838-848. [PMID: 37758602 DOI: 10.1016/j.tem.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, influencing numerous regulatory axes and extrahepatic vital organs. The molecular mechanisms that lead to the progression of NAFLD remain unclear and knowledge on the pathways causing hepatocellular damage followed by lipid accumulation is limited. Recently, a number of studies have shown that mRNA N6-methyladenosine (m6A) modification contributes to the progression of NAFLD. In this review, we summarize current knowledge on m6A modification in the metabolic processes associated with NAFLD and discuss the challenges of and prospects for therapeutic avenues based on m6A regulation for the treatment of liver disease.
Collapse
Affiliation(s)
- Ping Luo
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shiqi Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Jing
- Department of Clinical Laboratory, First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, China
| | - Jiancheng Tu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinghua Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
14
|
Ren Y, Li Z, Li J, Liang R, Wang Z, Bai Y, Yang Y, Tang Q, Fu Y, Zhang X, Zhang Y, Yu Y, Xiong Y. m 6 A mRNA methylation: Biological features, mechanisms, and therapeutic potentials in type 2 diabetes mellitus. Obes Rev 2023; 24:e13639. [PMID: 37732463 DOI: 10.1111/obr.13639] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/10/2023] [Accepted: 08/27/2023] [Indexed: 09/22/2023]
Abstract
As the most common internal post-transcriptional RNA modification in eukaryotic cells, N6-methyladenosine (m6 A) performs a dynamic and reversible role in a variety of biological processes mediated by methyltransferases (writers), demethylases (erasers), and m6 A binding proteins (readers). M6 A methylation enables transcriptome conversion in different signals that regulate various physiological activities and organ development. Over the past few years, emerging studies have identified that mRNA m6 A regulators defect in β-cell leads to abnormal regulation of the target mRNAs, thereby resulting in β-cell dysfunction and loss of β-cell identity and mass, which are strongly associated with type 2 diabetes mellitus (T2DM) pathogenesis. Also, mRNA m6 A modification has been implicated with insulin resistance in muscles, fat, and liver cells/tissues. In this review, we elaborate on the biological features of m6 A methylation; provide a comprehensive overview of the underlying mechanisms that how it controls β-cell function, identity, and mass as well as insulin resistance; highlight its connections to glucose metabolism and lipid metabolism linking to T2DM; and further discuss its role in diabetes complications and its therapeutic potentials for T2DM diagnosis and treatment.
Collapse
Affiliation(s)
- Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zi Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jiaoyu Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Rui Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yiduo Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yafang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Qian Tang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yaolei Fu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xiaobo Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yu Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- School of Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Rao G, Peng X, Li X, An K, He H, Fu X, Li S, An Z. Unmasking the enigma of lipid metabolism in metabolic dysfunction-associated steatotic liver disease: from mechanism to the clinic. Front Med (Lausanne) 2023; 10:1294267. [PMID: 38089874 PMCID: PMC10711211 DOI: 10.3389/fmed.2023.1294267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 06/21/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly defined as non-alcoholic fatty liver disease (NAFLD), is a disorder marked by the excessive deposition of lipids in the liver, giving rise to a spectrum of liver pathologies encompassing steatohepatitis, fibrosis/cirrhosis, and hepatocellular carcinoma. Despite the alarming increase in its prevalence, the US Food and Drug Administration has yet to approve effective pharmacological therapeutics for clinical use. MASLD is characterized by the accretion of lipids within the hepatic system, arising from a disarray in lipid provision (whether through the absorption of circulating lipids or de novo lipogenesis) and lipid elimination (via free fatty acid oxidation or the secretion of triglyceride-rich lipoproteins). This disarray leads to the accumulation of lipotoxic substances, cellular pressure, damage, and fibrosis. Indeed, the regulation of the lipid metabolism pathway is intricate and multifaceted, involving a myriad of factors, such as membrane transport proteins, metabolic enzymes, and transcription factors. Here, we will review the existing literature on the key process of lipid metabolism in MASLD to understand the latest progress in this molecular mechanism. Notably, de novo lipogenesis and the roles of its two main transcription factors and other key metabolic enzymes are highlighted. Furthermore, we will delve into the realm of drug research, examining the recent progress made in understanding lipid metabolism in MASLD. Additionally, we will outline prospective avenues for future drug research on MASLD based on our unique perspectives.
Collapse
Affiliation(s)
- Guocheng Rao
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Peng
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology and Metabolism, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, China
| | - Xinqiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Kang An
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, Multimorbidity Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - He He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shuangqing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, Multimorbidity Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Huang C, Chen W, Wang X. Studies on the fat mass and obesity-associated (FTO) gene and its impact on obesity-associated diseases. Genes Dis 2023; 10:2351-2365. [PMID: 37554175 PMCID: PMC10404889 DOI: 10.1016/j.gendis.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022] Open
Abstract
Obesity has become a major health crisis in the past ∼50 years. The fat mass and obesity-associated (FTO) gene, identified by genome-wide association studies (GWAS), was first reported to be positively associated with obesity in humans. Mice with more copies of the FTO gene were observed to be obese, while loss of the gene in mice was found to protect from obesity. Later, FTO was found to encode an m6A RNA demethylase and has a profound effect on many biological and metabolic processes. In this review, we first summarize recent studies that demonstrate the critical roles and regulatory mechanisms of FTO in obesity and metabolic disease. Second, we discuss the ongoing debates concerning the association between FTO polymorphisms and obesity. Third, since several small molecule drugs and micronutrients have been found to regulate metabolic homeostasis through controlling the expression or activity of FTO, we highlight the broad potential of targeting FTO for obesity treatment. Improving our understanding of FTO and the underlying mechanisms may provide new approaches for treating obesity and metabolic diseases.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Wei Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
17
|
Li Q, Wang W, Duan F, Wang Y, Chen S, Shi K, Xia Y, Li X, Gao Y, Liu G. DNMT3B Alleviates Liver Steatosis Induced by Chronic Low-grade LPS via Inhibiting CIDEA Expression. Cell Mol Gastroenterol Hepatol 2023; 17:59-77. [PMID: 37703946 PMCID: PMC10665944 DOI: 10.1016/j.jcmgh.2023.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease is the most prevalent chronic liver disease and threats to human health. Gut dysbiosis caused by lipopolysaccharide (LPS) leakage has been strongly related to nonalcoholic fatty liver disease progression, although the underlying mechanisms remain unclear. METHODS Previous studies have shown that low-grade LPS administration to mice on a standard, low-fat chow diet is sufficient to induce symptoms of fatty liver. This study confirmed these findings and supported LPS as a lipid metabolism regulator in the liver. RESULTS Mechanically, LPS induced dysregulated lipid metabolism by inhibiting the expression of DNA methyltransferases 3B (DNMT3B). Genetic overexpression of DNMT3B alleviated LPS-induced lipid accumulation, whereas its knockdown increased steatosis in mice and human hepatocytes. LPS-induced lower expression of DNMT3B led to hypomethylation in promoter region of CIDEA, resulting in increased binding of SREBP-1c to its promoter and activated CIDEA expression. Hepatic interference of CIDEA reversed the effect of LPS on lipogenesis. These effects were independent of a high-fat diet or high fatty acid action. CONCLUSIONS Overall, these findings sustain the conclusion that LPS is a lipogenic factor and could be involved in hepatic steatosis progression.
Collapse
Affiliation(s)
- Qiang Li
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China; Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui, China.
| | - Wenjing Wang
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Feifan Duan
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Yaju Wang
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Shuya Chen
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Kangyun Shi
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Yinyin Xia
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Xinyu Li
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Yu Gao
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China; Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Guoquan Liu
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui, China; Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China.
| |
Collapse
|
18
|
Song Y, Wade H, Zhang B, Xu W, Wu R, Li S, Su Q. Polymorphisms of Fat Mass and Obesity-Associated Gene in the Pathogenesis of Child and Adolescent Metabolic Syndrome. Nutrients 2023; 15:2643. [PMID: 37375547 DOI: 10.3390/nu15122643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Childhood metabolic syndrome (MetS) is prevalent around the world and is associated with a high likelihood of suffering from severe diseases such as cardiovascular disease later in adulthood. MetS is associated with genetic susceptibility that involves gene polymorphisms. The fat mass and obesity-associated gene (FTO) encodes an RNA N6-methyladenosine demethylase that regulates RNA stability and molecular functions. Human FTO contains genetic variants that significantly contribute to the early onset of MetS in children and adolescents. Emerging evidence has also uncovered that FTO polymorphisms in intron 1, such as rs9939609 and rs9930506 polymorphisms, are significantly associated with the development of MetS in children and adolescents. Mechanistic studies reported that FTO polymorphisms lead to aberrant expressions of FTO and the adjacent genes that promote adipogenesis and appetite and reduce steatolysis, satiety, and energy expenditure in the carriers. The present review highlights the recent observations on the key FTO polymorphisms that are associated with child and adolescent MetS with an exploration of the molecular mechanisms underlying the development of increased waist circumference, hypertension, and hyperlipidemia in child and adolescent MetS.
Collapse
Affiliation(s)
- Yongyan Song
- Central Laboratory, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu 610106, China
| | - Henry Wade
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast BT9 5DL, UK
| | - Bingrui Zhang
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast BT9 5DL, UK
| | - Wenhao Xu
- Clinical Medical College, Chengdu University, Chengdu 610106, China
| | - Rongxue Wu
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Shujin Li
- Central Laboratory, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu 610106, China
| | - Qiaozhu Su
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
19
|
Zhu X, Zhou C, Zhao S, Zheng Z. Role of m6A methylation in retinal diseases. Exp Eye Res 2023; 231:109489. [PMID: 37084873 DOI: 10.1016/j.exer.2023.109489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/06/2023] [Accepted: 04/19/2023] [Indexed: 04/23/2023]
Abstract
Retinal diseases remain among the leading causes of visual impairment in developed countries, despite great efforts in prevention and early intervention. Due to the limited efficacy of current retinal therapies, novel therapeutic methods are urgently required. Over the past two decades, advances in next-generation sequencing technology have facilitated research on RNA modifications, which can elucidate the relevance of epigenetic mechanisms to disease. N6-methyladenosine (m6A), formed by methylation of adenosine at the N6-position, is the most widely studied RNA modification and plays an important role in RNA metabolism. It is dynamically regulated by writers (methyltransferases) and erasers (demethylases), and recognized by readers (m6A binding proteins). Although the discovery of m6A methylation can be traced back to the 1970s, its regulatory roles in retinal diseases are rarely appreciated. Here, we provide an overview of m6A methylation, and discuss its effects and possible mechanisms on retinal diseases, including diabetic retinopathy, age-related macular degeneration, retinoblastoma, retinitis pigmentosa, and proliferative vitreoretinopathy. Furthermore, we highlight potential agents targeting m6A methylation for retinal disease treatment and discuss the limitations and challenges of research in the field of m6A methylation.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Chuandi Zhou
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Shuzhi Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| |
Collapse
|
20
|
Recent updates on targeting the molecular mediators of NAFLD. J Mol Med (Berl) 2023; 101:101-124. [PMID: 36792729 DOI: 10.1007/s00109-022-02282-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/29/2022] [Accepted: 12/21/2022] [Indexed: 02/17/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is rapidly becoming the most common disease worldwide in an era of rapid economic growth. NAFLD is a multifactorial disease, involving multiple genetic, metabolic, and environmental factors, and is closely associated with metabolic syndrome, obesity, and cardiovascular disease. NAFLD can be classified into nonalcoholic fatty liver disease (NAFL) and nonalcoholic steatohepatitis (NASH), which can both progress to cirrhosis and even hepatocellular carcinoma (HCC). Due to the enormous burden of NAFLD and its complications, no FDA-approved drugs for the treatment of NAFLD are on the market, and therapeutic targets and drug therapies are being actively investigated. In view of the various pathological mechanisms of NAFLD, numbers of preclinical studies and clinical trials have made rapid progress. This review mainly summarizes the most recently characterized mechanisms and therapeutic targets in each mechanism of NAFLD, focusing on the mechanism and application potential.
Collapse
|
21
|
Tan J, Wang YF, Dai ZH, Yin HZ, Mu CY, Wang SJ, Yang F. Roles of RNA m6A modification in nonalcoholic fatty liver disease. Hepatol Commun 2023; 7:e0046. [PMID: 38345896 PMCID: PMC9988276 DOI: 10.1097/hc9.0000000000000046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/08/2022] [Indexed: 02/15/2024] Open
Abstract
NAFLD is a series of liver disorders, and it has become the most prevalent hepatic disease to date. However, there are no approved and effective pharmaceuticals for NAFLD owing to a poor understanding of its pathological mechanisms. While emerging studies have demonstrated that m6A modification is highly associated with NAFLD. In this review, we summarize the general profile of NAFLD and m6A modification, and the role of m6A regulators including erasers, writers, and readers in NAFLD. Finally, we also highlight the clinical significance of m6A in NAFLD.
Collapse
Affiliation(s)
- Jian Tan
- The Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Yue-fan Wang
- The Department of Medical Genetics, Naval Medical University, Shanghai, China
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Zhi-hui Dai
- The Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Hao-zan Yin
- The Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Chen-yang Mu
- The Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Si-jie Wang
- The Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, Shanghai, China
| |
Collapse
|
22
|
Zhang N, Tian X, Yan T, Wang H, Zhang D, Lin C, Liu Q, Jiang S. Insights into the role of nucleotide methylation in metabolic-associated fatty liver disease. Front Immunol 2023; 14:1148722. [PMID: 37020540 PMCID: PMC10067741 DOI: 10.3389/fimmu.2023.1148722] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/22/2023] [Indexed: 04/07/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a chronic liver disease characterized by fatty infiltration of the liver. In recent years, the MAFLD incidence rate has risen and emerged as a serious public health concern. MAFLD typically progresses from the initial hepatocyte steatosis to steatohepatitis and then gradually advances to liver fibrosis, which may ultimately lead to cirrhosis and carcinogenesis. However, the potential evolutionary mechanisms still need to be clarified. Recent studies have shown that nucleotide methylation, which was directly associated with MAFLD's inflammatory grading, lipid synthesis, and oxidative stress, plays a crucial role in the occurrence and progression of MAFLD. In this review, we highlight the regulatory function and associated mechanisms of nucleotide methylation modification in the progress of MAFLD, with a particular emphasis on its regulatory role in the inflammation of MAFLD, including the regulation of inflammation-related immune and metabolic microenvironment. Additionally, we summarize the potential value of nucleotide methylation in the diagnosis and treatment of MAFLD, intending to provide references for the future investigation of MAFLD.
Collapse
Affiliation(s)
- Ni Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinchen Tian
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tinghao Yan
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haochen Wang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Dengtian Zhang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Cong Lin
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Qingbin Liu, ; Shulong Jiang,
| | - Shulong Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Qingbin Liu, ; Shulong Jiang,
| |
Collapse
|
23
|
Li L, Sun Y, Zha W, Li L, Li H. Novel insights into the N 6-methyladenosine RNA modification and phytochemical intervention in lipid metabolism. Toxicol Appl Pharmacol 2022; 457:116323. [PMID: 36427654 DOI: 10.1016/j.taap.2022.116323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/05/2022] [Accepted: 11/17/2022] [Indexed: 11/26/2022]
Abstract
Epitranscriptome (RNA modification) plays a vital role in a variety of biological events. N6-methyladenosine (m6A) modification is the most prevalent mRNA modification in eukaryotic cells. Dynamic and reversible m6A modification affects the plasticity of epitranscriptome, which plays an essential role in lipid metabolism. In this review, we comprehensively delineated the role and mechanism of m6A modification in the regulation of lipid metabolism in adipose tissue and liver, and summarized phytochemicals that improve lipid metabolism disturbance by targeting m6A regulator, providing potential lead candidates for drug therapeutics. Moreover, we discussed the main challenges and possible future directions in this field.
Collapse
Affiliation(s)
- Linghuan Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Yuanhai Sun
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Weiwei Zha
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Lingqing Li
- Taizhou Municipal Hospital, Taizhou University, Taizhou 318000, PR China
| | - Hanbing Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, PR China.
| |
Collapse
|
24
|
Interplay between the m 6A Epitranscriptome and Tumor Metabolism: Mechanisms and Therapeutic Implications. Biomedicines 2022; 10:biomedicines10102589. [PMID: 36289851 PMCID: PMC9599308 DOI: 10.3390/biomedicines10102589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
N6-methyladenosine (m6A) modification of messenger RNA (mRNA) influences the stability and translation of the transcripts into functional proteins. Recent studies reveal the role of m6A modifications in regulating the metabolism of basic biomolecules such as glucose, lipids and amino acids. Such mechanisms are not only important for physiological functions of normal cells but also prove to be pivotal for the pathogenesis of cancers by driving dysregulated metabolism. M6A writers, readers and erasers function co-operatively to promote aberrant glucose, lipid and amino acid metabolism in cancer cells, which in turn support increased proliferative and metastatic potential. Better understanding of the relationship between m6A and metabolism in malignancy may unravel novel therapeutic targets as well as biomarkers in cancer. In this review, we summarize the recent evidence demonstrating the interplay between m6A modification and cancer metabolism and their therapeutic implications.
Collapse
|
25
|
How to Use Respiratory Chain Inhibitors in Toxicology Studies-Whole-Cell Measurements. Int J Mol Sci 2022; 23:ijms23169076. [PMID: 36012337 PMCID: PMC9409450 DOI: 10.3390/ijms23169076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial electron transport chain (ETC) inhibition is a phenomenon interesting in itself and serves as a tool for studying various cellular processes. Despite the fact that searching the term “rotenone” in PubMed returns more than 6900 results, there are many discrepancies regarding the directions of changes reported to be caused by this RTC inhibitor in the delicate redox balance of the cell. Here, we performed a multifaceted study of the popular ETC inhibitors rotenone and antimycin A, involving assessment of mitochondrial membrane potential and the production of hydrogen peroxide and superoxide anions at cellular and mitochondrial levels over a wide range of inhibitor concentrations (1 nmol/dm3–100 µmol/dm3). All measurements were performed with whole cells, with accompanying control of ATP levels. Antimycin A was more potent in hindering HepG2 cells’ abilities to produce ATP, decreasing ATP levels even at a 1 nmol/dm3 concentration, while in the case of rotenone, a 10,000-times greater concentration was needed to produce a statistically significant decrease. The amount of hydrogen peroxide produced in the course of antimycin A biological activity increased rapidly at low concentrations and decreased below control level at a high concentration of 100 µmol/dm3. While both inhibitors influenced cellular superoxide anion production in a comparable manner, rotenone caused a greater increase in mitochondrial superoxide anions compared to a modest impact for antimycin A. IC50 values for rotenone and antimycin A with respect to HepG2 cell survival were of the same order of magnitude, but the survival curve of cells treated with rotenone was clearly biphasic, suggesting a concentration-dependent mode of biological action. We propose a clear experimental setup allowing for complete and credible analysis of the redox state of cells under stress conditions which allows for better understanding of the effects of ETC inhibition.
Collapse
|
26
|
Zhang F, Liu H, Duan M, Wang G, Zhang Z, Wang Y, Qian Y, Yang Z, Jiang X. Crosstalk among m6A RNA methylation, hypoxia and metabolic reprogramming in TME: from immunosuppressive microenvironment to clinical application. J Hematol Oncol 2022; 15:84. [PMID: 35794625 PMCID: PMC9258089 DOI: 10.1186/s13045-022-01304-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/09/2022] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME), which is regulated by intrinsic oncogenic mechanisms and epigenetic modifications, has become a research hotspot in recent years. Characteristic features of TME include hypoxia, metabolic dysregulation, and immunosuppression. One of the most common RNA modifications, N6-methyladenosine (m6A) methylation, is widely involved in the regulation of physiological and pathological processes, including tumor development. Compelling evidence indicates that m6A methylation regulates transcription and protein expression through shearing, export, translation, and processing, thereby participating in the dynamic evolution of TME. Specifically, m6A methylation-mediated adaptation to hypoxia, metabolic dysregulation, and phenotypic shift of immune cells synergistically promote the formation of an immunosuppressive TME that supports tumor proliferation and metastasis. In this review, we have focused on the involvement of m6A methylation in the dynamic evolution of tumor-adaptive TME and described the detailed mechanisms linking m6A methylation to change in tumor cell biological functions. In view of the collective data, we advocate treating TME as a complete ecosystem in which components crosstalk with each other to synergistically achieve tumor adaptive changes. Finally, we describe the potential utility of m6A methylation-targeted therapies and tumor immunotherapy in clinical applications and the challenges faced, with the aim of advancing m6A methylation research.
Collapse
|
27
|
Cai X, Liang C, Zhang M, Xu Y, Weng Y, Li X, Yu W. N6-methyladenosine modification and metabolic reprogramming of digestive system malignancies. Cancer Lett 2022; 544:215815. [DOI: 10.1016/j.canlet.2022.215815] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 12/12/2022]
|
28
|
Wei X, Zhang J, Tang M, Wang X, Fan N, Peng Y. Fat mass and obesity–associated protein promotes liver steatosis by targeting PPARα. Lipids Health Dis 2022; 21:29. [PMID: 35282837 PMCID: PMC8918283 DOI: 10.1186/s12944-022-01640-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/28/2022] [Indexed: 12/27/2022] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide. The fat mass and obesity–associated protein (FTO) has been shown to be involved in obesity; however, its role in NAFLD and the underlying molecular mechanisms remain largely unknown. Methods FTO expression was first examined in the livers of patients with NAFLD and animal and cellular models of NAFLD by real-time PCR and Western blotting. Next, its role in lipid accumulation in hepatocytes was assessed both in vitro and in vivo via gene overexpression and knockdown studies. Results FTO expression was obviously elevated in the livers of mice and humans with hepatic steatosis, probably due to its decreased ubiquitination. FTO overexpression in HepG2 cells induced triglyceride accumulation, whereas FTO knockdown exerted an opposing effect. Consistent with the findings of in vitro studies, adeno-associated viruses 8 (AAV8)-mediated FTO overexpression in the liver promoted hepatic steatosis in C57BL/6J mice. Mechanistically, FTO inhibited the mRNA of peroxisome proliferator-activated receptor α (PPARα) in hepatocytes. Activation of PPARα by its agonist GW7647 reversed lipid accumulation in hepatocytes induced by FTO overexpression. Conclusions Overall, FTO expression is increased in NAFLD, and it promotes hepatic steatosis by targeting PPARα. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01640-y.
Collapse
|
29
|
Wilkinson E, Cui YH, He YY. Roles of RNA Modifications in Diverse Cellular Functions. Front Cell Dev Biol 2022; 10:828683. [PMID: 35350378 PMCID: PMC8957929 DOI: 10.3389/fcell.2022.828683] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/14/2022] [Indexed: 12/19/2022] Open
Abstract
Chemical modifications of RNA molecules regulate both RNA metabolism and fate. The deposition and function of these modifications are mediated by the actions of writer, reader, and eraser proteins. At the cellular level, RNA modifications regulate several cellular processes including cell death, proliferation, senescence, differentiation, migration, metabolism, autophagy, the DNA damage response, and liquid-liquid phase separation. Emerging evidence demonstrates that RNA modifications play active roles in the physiology and etiology of multiple diseases due to their pervasive roles in cellular functions. Here, we will summarize recent advances in the regulatory and functional role of RNA modifications in these cellular functions, emphasizing the context-specific roles of RNA modifications in mammalian systems. As m6A is the best studied RNA modification in biological processes, this review will summarize the emerging advances on the diverse roles of m6A in cellular functions. In addition, we will also provide an overview for the cellular functions of other RNA modifications, including m5C and m1A. Furthermore, we will also discuss the roles of RNA modifications within the context of disease etiologies and highlight recent advances in the development of therapeutics that target RNA modifications. Elucidating these context-specific functions will increase our understanding of how these modifications become dysregulated during disease pathogenesis and may provide new opportunities for improving disease prevention and therapy by targeting these pathways.
Collapse
Affiliation(s)
- Emma Wilkinson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| | - Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, United States
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
30
|
Wang Y, Liu B, Wu P, Chu Y, Gui S, Zheng Y, Chen X. Dietary Selenium Alleviated Mouse Liver Oxidative Stress and NAFLD Induced by Obesity by Regulating the KEAP1/NRF2 Pathway. Antioxidants (Basel) 2022; 11:antiox11020349. [PMID: 35204232 PMCID: PMC8868436 DOI: 10.3390/antiox11020349] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/05/2022] [Accepted: 02/06/2022] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) occurs when excess fat is stored in the liver and it is strongly linked with metabolic syndrome and oxidative stress. Selenium (Se) is an essential micronutrient in animals, which has a variety of biological functions, including antioxidant and anti-inflammatory. However, the exact effect of dietary selenium on NAFLD and the underlying molecular mechanism are not yet clear. Herein, we fed a high-fat diet (HFD) to C57BL/6 mice to construct an in vivo NAFLD model, treated AML-12 cells with palmitic acid (PA) to construct an in vitro NAFLD model, and AML-12 cells were stimulated with H2O2 to induce hepatocyte oxidative stress and then treated with adequate selenium. We observed that adequate selenium significantly improved the hepatic injury and insulin resistance in HFD mice, and decreased the fat accumulation and the expression of lipogenic genes in PA-induced AML-12 cells. Meanwhile, selenium significantly inhibited the production of reactive oxygen species (ROS), inhibited apoptosis, and restored mitochondrial number and membrane potential in PA- induced AML-12 cells. In addition, selenium can promote selenoproteinP1 (SEPP1) synthesis to regulate the Kelch-like ECH-associated protein 1 (KEAP1)/NF-E2-related factor 2 (NRF2) pathway, so as to defend against hepatocyte oxidative stress. These findings suggest that dietary selenium supplementation can effectively resist hepatic injury and insulin resistance during NAFLD development, and regulate the KEAP1/NRF2 pathway to resist oxidative stress by promoting SEPP1 synthesis.
Collapse
Affiliation(s)
- Yi Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (B.L.); (Y.C.); (S.G.); (Y.Z.)
| | - Bingbing Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (B.L.); (Y.C.); (S.G.); (Y.Z.)
| | - Peixuan Wu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Yi Chu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (B.L.); (Y.C.); (S.G.); (Y.Z.)
| | - Sisi Gui
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (B.L.); (Y.C.); (S.G.); (Y.Z.)
| | - Yazhen Zheng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (B.L.); (Y.C.); (S.G.); (Y.Z.)
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.W.); (B.L.); (Y.C.); (S.G.); (Y.Z.)
- Correspondence: ; Tel.: +86-27-87282091
| |
Collapse
|
31
|
Zhou W, Cai D. Midazolam suppresses ischemia/reperfusion-induced cardiomyocyte apoptosis by inhibiting the JNK/p38 MAPK signaling pathway. Can J Physiol Pharmacol 2022; 100:117-124. [PMID: 34559975 DOI: 10.1139/cjpp-2021-0289] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury causes irreversible injury to the heart, thereby causing acute myocardial infarction. Midazolam is a benzodiazepine commonly utilized in anesthesia and intensive care. Research has indicated that midazolam plays a critical role in many diseases; however, the function of midazolam in myocardial injury induced by I/R still needs further investigation. The infarct size and damage to the heart tissues were examined through 2,3,5-triphenyl tetrazolium chloride (TTC) staining and hematoxylin and eosin staining. The creatine kinase-myocardial band isoenzyme, lactate dehydrogenase, and aspartate aminotransferase levels were tested using commercial kits. Cell apoptosis was determined through TUNEL staining or flow cytometry assays. Bax, Bcl-2, cleaved caspase-3, phospho-38 (p-p38), p38, p-JNK, JNK, extracellular signal-regulated kinases (ERK), and p-ERK expression was examined through Western blot. In our study, midazolam was shown to suppress the infarct size and heart tissue damage and reduce myocardial enzyme leakage in I/R rats. Additionally, midazolam was found to retard cardiomyocyte apoptosis in I/R rats. The JNK/p38 MAPK signaling pathway in I/R rats was inhibited by midazolam. Our findings demonstrated that in hypoxia/reoxygenation (H/R) - mediated H9C2 cells, anisomycin abolished the suppressive effects of midazolam on the JNK/p38 MAPK signaling pathway. Next, exploration discovered that anisomycin abolished the cytoprotective effects of midazolam on H/R-treated H9C2 cell apoptosis. In conclusion, this work demonstrated that midazolam retarded I/R-induced cardiomyocyte apoptosis by inhibiting the JNK/p38 MAPK signaling pathway. These results may provide new insight into the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Weixiao Zhou
- Department of Anesthesiology, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, Zhejiang 311800, China
- Department of Anesthesiology, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, Zhejiang 311800, China
| | - Dongjiang Cai
- Department of Anesthesiology, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, Zhejiang 311800, China
- Department of Anesthesiology, Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, Zhejiang 311800, China
| |
Collapse
|
32
|
Yang Z, Yu GL, Zhu X, Peng TH, Lv YC. Critical roles of FTO-mediated mRNA m6A demethylation in regulating adipogenesis and lipid metabolism: Implications in lipid metabolic disorders. Genes Dis 2022; 9:51-61. [PMID: 35005107 PMCID: PMC8720706 DOI: 10.1016/j.gendis.2021.01.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/19/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
The goal this review is to clarify the effects of the fat mass and obesity-associated protein (FTO) in lipid metabolism regulation and related underlying mechanisms through the FTO-mediated demethylation of m6A modification. FTO catalyzes the demethylation of m6A to alter the processing, maturation and translation of the mRNAs of lipid-related genes. FTO overexpression in the liver promotes lipogenesis and lipid droplet (LD) enlargement and suppresses CPT-1–mediated fatty acid oxidation via the SREBP1c pathway, promoting excessive lipid storage and nonalcoholic fatty liver diseases (NAFLD). FTO enhances preadipocyte differentiation through the C/EBPβ pathway, and facilitates adipogenesis and fat deposition by altering the alternative splicing of RUNX1T1, the expression of PPARγ and ANGPTL4, and the phosphorylation of PLIN1, whereas it inhibits lipolysis by inhibiting IRX3 expression and the leptin pathway, causing the occurrence and development of obesity. Suppression of the PPARβ/δ and AMPK pathways by FTO-mediated m6A demethylation damages lipid utilization in skeletal muscles, leading to the occurrence of diabetic hyperlipidemia. m6A demethylation by FTO inhibits macrophage lipid influx by downregulating PPARγ protein expression and accelerates cholesterol efflux by phosphorylating AMPK, thereby impeding foam cell formation and atherosclerosis development. In summary, FTO-mediated m6A demethylation modulates the expression of lipid-related genes to regulate lipid metabolism and lipid disorder diseases.
Collapse
Affiliation(s)
- Zhou Yang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Guang-Li Yu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Tian-Hong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Yun-Cheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.,Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| |
Collapse
|
33
|
Choubey P, Kaur H, Bansal K. Modulation of DNA/RNA Methylation Signaling Mediating Metabolic Homeostasis in Cancer. Subcell Biochem 2022; 100:201-237. [PMID: 36301496 DOI: 10.1007/978-3-031-07634-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Nucleic acid methylation is a fundamental epigenetic mechanism that impinges upon several cellular attributes, including metabolism and energy production. The dysregulation of deoxyribonucleic acid (DNA)/ribonucleic acid (RNA) methylation can lead to metabolic rewiring in the cell, which in turn facilitates tumor development. Here, we review the current knowledge on the interplay between DNA/RNA methylation and metabolic programs in cancer cells. We also discuss the mechanistic role of these pathways in tumor development and progression.
Collapse
Affiliation(s)
- Pallawi Choubey
- Molecular Biology and Genetics Unit (MBGU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore, India
| | - Harshdeep Kaur
- Molecular Biology and Genetics Unit (MBGU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore, India
| | - Kushagra Bansal
- Molecular Biology and Genetics Unit (MBGU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore, India.
| |
Collapse
|
34
|
Wang Y, Wang Y, Gu J, Su T, Gu X, Feng Y. The role of RNA m6A methylation in lipid metabolism. Front Endocrinol (Lausanne) 2022; 13:866116. [PMID: 36157445 PMCID: PMC9492936 DOI: 10.3389/fendo.2022.866116] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
The m6A methylation is the most numerous modification of mRNA in mammals, coordinated by RNA m6A methyltransferases, RNA m6A demethylases, and RNA m6A binding proteins. They change the RNA m6A methylation level in their specific manner. RNA m6A modification has a significant impact on lipid metabolic regulation. The "writer" METTL3/METTL14 and the "eraser" FTO can promote the accumulation of lipids in various cells by affecting the decomposition and synthesis of lipids. The "reader" YTHDF recognizes m6A methylation sites of RNA and regulates the target genes' translation. Due to this function that regulates lipid metabolism, RNA m6A methylation plays a pivotal role in metabolic diseases and makes it a great potential target for therapy.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yujie Wang
- Department of Orthopaedics, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Jiarui Gu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tianhong Su
- Department of Cardiology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaosong Gu
- Department of Cardiology, the Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Yu Feng, ; Xiaosong Gu,
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Yu Feng, ; Xiaosong Gu,
| |
Collapse
|
35
|
Wei M, Zhan D, Li ZX, Wang HY, Xing Y, Luo XP. Effect of high-fat diet for rats at different stages on glucose and lipid metabolism in offspring and related mechanisms. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:1174-1183. [PMID: 34753551 DOI: 10.7499/j.issn.1008-8830.2107121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To study the effect of high-fat diet for maternal Sprague-Dawley rats at different stages on glucose and lipid metabolism in offspring and related mechanisms. METHODS According to the diet before pregnancy and during pregnancy and lactation, maternal rats were randomly divided into four groups (n=9 each): CC (control diet before pregnancy and during pregnancy and lactation), HC (high-fat diet before pregnancy and control diet during pregnancy and lactation), CH (control diet before pregnancy and high-fat diet during pregnancy and lactation), and HH (high-fat diet before pregnancy and during pregnancy and lactation), and all offspring rats were given control diet after weaning. The body weight of maternal rats was recorded before and during pregnancy. Male offspring rats were selected from each group at the juvenile stage (3-week old) and the adult stage (12-week old) to measure the levels of fasting blood glucose (FBG) and fasting insulin (FINS) and the levels of triglyceride (TG) and total cholesterol (TC) in the liver. Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) index was calculated, and the area under the curve (AUC) was calculated for glucose tolerance test (GTT) and insulin tolerance test (ITT). Lipid deposition in the liver was observed, and the mRNA and protein expression levels of the key genes in glucose and lipid metabolism (IR, IRS, and AKT), FASN, SREBP1c, and PPARα in the liver were also measured. RESULTS Compared with the control diet groups (CC and CH groups), the groups with high-fat diet before pregnancy (HC and HH groups) had a significant increase in body weight (P<0.001). Compared with the CC group, the HC, CH, and HH groups had a significantly greater increase in body weight (P<0.001). Compared with the CC group, the HC, CH, and HH groups had significant increases in body weight, the levels of TG and TC in the liver, and the mRNA and protein expression levels of FASN, SREBP1c, and PPARα in the offspring rats at week 3 after birth (P<0.05), as well as a significant increase in lipid deposition in the liver, with the most significant increase of the parameters in the HH group. Compared with the CC group, the HH group had significant increases in the levels of FBG and FINS, HOMA-IR index, GTT-AUC, ITT-AUC, and the protein expression level of p-IRS in the liver and significant reductions in the mRNA and protein expression levels of IR and IRS in the liver in the offspring rats at week 3 after birth (P<0.05). Compared with the CC group, the HC, CH, and HH groups had significant increases in body weight, the levels of FBG and FINS, HOMA-IR index, GTT-AUC, ITT-AUC, the levels of TG and TC in the liver, protein expression level of p-IRS in the liver, and the mRNA and protein expression levels of FASN, SREBP1c, and PPARα in the offspring rats at week 12 after birth (P<0.05), as well as a significant increase in lipid deposition in the liver, with the most increase of the parameters in the HH group. Compared with the CC group, the HC, CH, and HH groups had significant reductions in the mRNA expression levels of IR, IRS, and AKT and the protein expression levels of IR, IRS, and p-AKT in the offspring rats at week 12 after birth (P<0.05). There were no significant differences in the levels of glucose and lipid metabolism between the HC and CH groups at various stages (P>0.05). CONCLUSIONS High-fat diet for rats at different stages before and after pregnancy has different effects on glucose and lipid metabolism of offspring rats, and high-fat diet before pregnancy and during pregnancy and lactation has the greatest effect. The effect of high-fat diet on glucose and lipid metabolism of offspring rats is considered associated with the changes in the expression of genes involved in glucose and lipid metabolism.
Collapse
Affiliation(s)
- Ming Wei
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Di Zhan
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhu-Xi Li
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huan-Yu Wang
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Xing
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Ping Luo
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
36
|
Cheng L, Yu P, Li F, Jiang X, Jiao X, Shen Y, Lai X. Human umbilical cord-derived mesenchymal stem cell-exosomal miR-627-5p ameliorates non-alcoholic fatty liver disease by repressing FTO expression. Hum Cell 2021; 34:1697-1708. [PMID: 34410623 DOI: 10.1007/s13577-021-00593-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/11/2021] [Indexed: 01/17/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of liver disorders. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs)-based therapy is currently considered to be an effective treatment for NAFLD. The present study aimed to determine whether hUC-MSCs-exosomes have a hepatoprotective effect on NAFLD. We constructed NAFLD rat model by high-fat high-fructose feeding. Liver cells (L-O2) were treated with palmitic acid (PA) to mimic NAFLD model. NAFLD rats and PA-treated L-O2 cells were treated with hUC-MSCs-exosomes, and then we determined the influence of exosomes on liver damage and glucose and lipid metabolism in vivo and in vitro. We found that hUC-MSCs-exosomes exhibited an up-regulation of miR-627-5p. Exosomal miR-627-5p promoted cell viability and repressed apoptosis of PA-treated L-O2 cells. Exosomal miR-627-5p also enhanced the expression of G6Pc, PEPCK, FAS and SREBP-1c and suppressed PPARα expression in PA-treated L-O2 cells. Moreover, miR-627-5p interacted with fat mass and obesity-associated gene (FTO) and inhibited FTO expression in L-O2 cells. MiR-627-5p-enriched exosomes improved glucose and lipid metabolism in L-O2 cells by targeting FTO. In vivo, exosomal miR-627-5p ameliorated insulin tolerance, liver damage, glucose and lipid metabolism and reduced lipid deposition in NAFLD rats. Exosomal miR-627-5p also reduced body weight, liver weight, and liver index (body weight/liver weight) in NAFLD rats. In conclusion, these data demonstrate that HUC-MSCs-derived exosomal miR-627-5p improves glucose and lipid metabolism and alleviate liver damage by repressing FTO expression, thereby ameliorating NAFLD progression. Thus, hUC-MSCs-exosomes may be a potential treatment for NAFLD.
Collapse
Affiliation(s)
- Lidan Cheng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Fangfang Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Xueling Jiang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Xiaojuan Jiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China.
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China.
| | - Xiaoyang Lai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China.
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China.
| |
Collapse
|
37
|
Regulatory role and mechanism of m 6A RNA modification in human metabolic diseases. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:52-63. [PMID: 34485686 PMCID: PMC8399361 DOI: 10.1016/j.omto.2021.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metabolic diseases caused by disorders in amino acids, glucose, lipid metabolism, and other metabolic risk factors show high incidences in young people, and current treatments are ineffective. N6-methyladenosine (m6A) RNA modification is a post-transcriptional regulation of gene expression with several effects on physiological processes and biological functions. Recent studies report that m6A RNA modification is involved in various metabolic pathways and development of common metabolic diseases, making it a potential disease-specific therapeutic target. This review explores components, mechanisms, and research methods of m6A RNA modification. In addition, we summarize the progress of research on m6A RNA modification in metabolism-related human diseases, including diabetes, obesity, non-alcoholic fatty liver disease, osteoporosis, and cancer. Furthermore, opportunities and the challenges facing basic research and clinical application of m6A RNA modification in metabolism-related human diseases are discussed. This review is meant to enhance our understanding of the molecular mechanisms, research methods, and clinical significance of m6A RNA modification in metabolism-related human diseases.
Collapse
|
38
|
Sun D, Zhao T, Zhang Q, Wu M, Zhang Z. Fat mass and obesity-associated protein regulates lipogenesis via m 6 A modification in fatty acid synthase mRNA. Cell Biol Int 2020; 45:334-344. [PMID: 33079435 DOI: 10.1002/cbin.11490] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/01/2020] [Accepted: 10/18/2020] [Indexed: 12/13/2022]
Abstract
As the first identified N6 -methyladenosine (m6 A) demethylase, fat mass and obesity-associated (FTO) protein is associated with fatty acid synthase (FASN) and lipid accumulation. However, little is known about the regulatory role of FTO in the expression of FASN and de novo lipogenesis through m6 A modification. In this study, we used FTO small interfering RNA to explore the effects of FTO knockdown on hepatic lipogenesis and its underlying epigenetic mechanism in HepG2 cells. We found that knockdown of FTO increased m6 A levels in total RNA and enhanced the expression of YTH domain family member 2 which serves as the m6 A-binding protein. The de novo lipogenic enzymes and intracellular lipid content were significantly decreased under FTO knockdown. Mechanistically, knockdown of FTO dramatically enhanced m6 A levels in FASN messenger RNA (mRNA), leading to the reduced expression of FASN mRNA through m6 A-mediated mRNA decay. The protein expressions of FASN along with acetyl CoA carboxylase and ATP-citrate lyase were further decreased, which inhibited de novo lipogenesis, thereby resulting in the deficiency of lipid accumulation in HepG2 cells and the induction of cellular apoptosis. The results reveal that FTO regulates hepatic lipogenesis via FTO-dependent m6 A demethylation in FASN mRNA and indicate the critical role of FTO-mediated lipid metabolism in the survival of HepG2 cells. This study provides novel insights into a unique RNA epigenetic mechanism by which FTO mediates hepatic lipid accumulation through m6 A modification and indicates that FTO could be a potential target for obesity-related diseases and cancer.
Collapse
Affiliation(s)
- Donglei Sun
- Department of Environmental and Occupational Health, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianhe Zhao
- Department of Environmental and Occupational Health, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhang
- Department of Environmental and Occupational Health, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mei Wu
- Department of Environmental and Occupational Health, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zunzhen Zhang
- Department of Environmental and Occupational Health, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Cheng J, Liu Y, Liu Y, Liu D, Liu Y, Guo Y, Wu Z, Li H, Wang H. Ursolic acid alleviates lipid accumulation by activating the AMPK signaling pathway in vivo and in vitro. J Food Sci 2020; 85:3998-4008. [PMID: 33001454 DOI: 10.1111/1750-3841.15475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/10/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
Abstract
The mechanism underlying the effect of ursolic acid (UA) on lipid metabolism remains unclear. This study aimed to explore the mechanisms of UA in reducing lipid accumulation in free fatty acids-cultured HepG2 cells and in high-fat-diet-fed C57BL/6J mice. In vivo, UA effectively alleviated liver steatosis and decreased the size of adipocytes in the epididymis. It also significantly decreased the total cholesterol (TC) and triglyceride (TG) contents in the liver and plasma in C57BL/6 mice. In vitro, UA (20 µM) significantly reduced lipid accumulation; the intracellular TC contents decreased from 0.078 ± 0.0047 to 0.049 ± 0.0064 µmol/mg protein, and TG contents from 0.133 ± 0.005 to 0.066 ± 0.0047 µmol/mg protein, in HepG2 cells. Furthermore, UA reduced the mRNA expression related to fat synthesis, enhanced the mRNA expression related to adipose decomposition, and dramatically upregulated the protein expression of P-AMPK in vivo and in vitro. Of note, these protective effects of UA on a high-fat environment were blocked by the AMPK inhibitor (compound C) in vitro. In addition, the molecular docking results suggested that UA could be docked to the AMPK protein as an AMPK activator. These results indicated that UA lowered the lipid content probably via activating the AMPK signaling pathway, thereby inhibiting lipid synthesis and promoting fat decomposition. PRACTICAL APPLICATION: Ursolic acid (UA) widely exists in vegetables and fruits. This study highlighted a lipid-lowing mechanism of UA in HepG2 cells and C57BL/6J mice. The data indicated that UA might be used in lipid-lowering functional foods.
Collapse
Affiliation(s)
- Jing Cheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Ying Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Yaojie Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Dong Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| | - Yang Liu
- Animal and Plant and Food Inspection Center of Tianjin Customs (Former Tianjin Inspection and Quarantine Bureau), Tianjin, 300461, China
| | - Yatu Guo
- Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, 300384, China
| | - Zijian Wu
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300143, China
| | - Heyu Li
- Tianjin Ubasio Biotechnology Group Co. Ltd., Tianjin, 300457, China
| | - Hao Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, 300457, China
| |
Collapse
|
40
|
Han X, Wang L, Han Q. Advances in the role of m 6A RNA modification in cancer metabolic reprogramming. Cell Biosci 2020; 10:117. [PMID: 33062255 PMCID: PMC7552565 DOI: 10.1186/s13578-020-00479-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/24/2020] [Indexed: 01/10/2023] Open
Abstract
N6-methyladenosine (m6A) modification is the most common internal modification of eukaryotic mRNA and is widely involved in many cellular processes, such as RNA transcription, splicing, nuclear transport, degradation, and translation. m6A has been shown to plays important roles in the initiation and progression of various cancers. The altered metabolic programming of cancer cells promotes their cell-autonomous proliferation and survival, leading to an indispensable hallmark of cancers. Accumulating evidence has demonstrated that this epigenetic modification exerts extensive effects on the cancer metabolic network by either directly regulating the expression of metabolic genes or modulating metabolism-associated signaling pathways. In this review, we summarized the regulatory mechanisms and biological functions of m6A and its role in cancer metabolic reprogramming.
Collapse
Affiliation(s)
- Xiu Han
- Center of Clinical Laboratory, Suzhou Dushu Lake Public Hospital, 9#, Chongwen Road, Suzhou, 215000 People’s Republic of China
| | - Lin Wang
- Center of Clinical Laboratory, Suzhou Dushu Lake Public Hospital, 9#, Chongwen Road, Suzhou, 215000 People’s Republic of China
| | - Qingzhen Han
- Center of Clinical Laboratory, Suzhou Dushu Lake Public Hospital, 9#, Chongwen Road, Suzhou, 215000 People’s Republic of China
| |
Collapse
|
41
|
Zhao Z, Meng J, Su R, Zhang J, Chen J, Ma X, Xia Q. Epitranscriptomics in liver disease: Basic concepts and therapeutic potential. J Hepatol 2020; 73:664-679. [PMID: 32330603 DOI: 10.1016/j.jhep.2020.04.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
The development of next-generation sequencing technology and the discovery of specific antibodies targeting chemically modified nucleotides have paved the way for a new era of epitranscriptomics. Cellular RNA is known to dynamically and reversibly undergo different chemical modifications after transcription, such as N6-methyladenosine (m6A), N1-methyladenosine, N6,2'-O-dimethyladenosine, 5-methylcytosine, and 5-hydroxymethylcytidine, whose identity and location comprise the field of epitranscriptomics. Dynamic post-transcriptional modifications determine the fate of target RNAs by regulating various aspects of their processing, including RNA export, transcript processing, splicing, and degradation. The most abundant internal mRNA modification in eukaryotic cells is m6A, which exhibits essential roles in physiological processes, such as embryogenesis, carcinogenesis, and neurogenesis. m6A is deposited by the m6A methyltransferase complex (composed of METTL3/14/16, WTAP, KIAA1429, and RBM15/15B), erased by demethylases (FTO and ALKBH5), and recognised by binding proteins (e.g., YTHDF1/2/3, YTHDC1/2, IGF2BP1/2/3). The liver is the largest digestive and metabolic organ, and m6A modifications play unique roles in critical physiological hepatic functions and various liver diseases. This review focuses on the biological roles of m6A RNA methylation in lipid metabolism, viral hepatitis, non-alcoholic fatty liver disease, liver cancer, and tumour metastasis. In addition, we summarise the existing inhibitors targeting m6A regulators and discuss the potential of modulating m6A modifications as a therapeutic strategy.
Collapse
Affiliation(s)
- Zhicong Zhao
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Jiaxiang Meng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Jun Zhang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai 200001, China
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai 200001, China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
42
|
Zhang L, Ding L, Shi H, Wang C, Xue C, Zhang T, Wang Y. Eicosapentaenoic acid-enriched phospholipids suppressed lipid accumulation by specific inhibition of lipid droplet-associated protein FSP27 in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:2244-2251. [PMID: 31919850 DOI: 10.1002/jsfa.10250] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/22/2019] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Sea cucumber is a rich source of eicosapentaenoic acid in the form of eicosapentaenoic acid-enriched phospholipids (EPA-PL). It is known to be efficacious in preventing obesity. However, few studies have focused on the role of EPA-PL in inhibiting lipid accumulation by lipid droplets (LDs). This study first investigated the effect of EPA-PL from sea cucumber on the formation of LDs and the underlying mechanism in C57BL/6J mice. The mice were randomly divided into two groups and treated for 8 weeks or 3, 7, and 14 days with either (i) a high-sucrose diet (model group), (ii) a high-sucrose diet plus 2% EPA-PL (EPA-PL group). RESULTS Eight-week EPA-PL supplementation significantly reduced lipid accumulation and LD size in liver and white adipose tissue (WAT), which was accompanied by the decreased expression of LDs-associated protein FSP27. A 3-day EPA-PL treatment suppressed the mRNA expression of Fsp27. The mRNA level of Fsp27 reached its 'normal level' after withdrawing EPA-PL for 7 days, suggesting that EPA-PL might serve as a rapid regulator of FSP27. Furthermore, EPA-PL increased the expression of lipolysis genes Hsl and Atgl accompanied by the regulation of Pparγ in WAT. CONCLUSIONS Dietary EPA-PL from sea cucumber (Cucumaria frondosa) protected against lipid accumulation by regulating LDs-associated protein FSP27, which might provide novel evidence for the anti-obesity action of EPA-PL. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Lingyu Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Lin Ding
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Haohao Shi
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Chengcheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological Products, Qingdao, China
| | - Tiantian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yuming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological Products, Qingdao, China
| |
Collapse
|
43
|
Brulport A, Vaiman D, Chagnon MC, Le Corre L. Obesogen effect of bisphenol S alters mRNA expression and DNA methylation profiling in male mouse liver. CHEMOSPHERE 2020; 241:125092. [PMID: 31683443 DOI: 10.1016/j.chemosphere.2019.125092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
Environmental pollution is increasingly considered an important factor involved in the obesity incidence. Endocrine disruptors (EDs) are important actors in the concept of DOHaD (Developmental Origins of Health and Disease), where epigenetic mechanisms play crucial roles. Bisphenol A (BPA), a monomer used in the manufacture of plastics and resins is one of the most studied obesogenic endocrine disruptor. Bisphenol S (BPS), a BPA substitute, has the same obesogenic properties, acting at low doses with a sex-specific effect following perinatal exposure. Since the liver is a major organ in regulating body lipid homeostasis, we investigated gene expression and DNA methylation under low-dose BPS exposure. The BPS obesogenic effect was associated with an increase of hepatic triglyceride content. These physiological disturbances were accompanied by genome-wide changes in gene expression (1366 genes significantly modified more than 1.5-fold). Gene ontology analysis revealed alteration of gene cascades involved in protein translation and complement regulation. It was associated with hepatic DNA hypomethylation in autosomes and hypermethylation in sex chromosomes. Although no systematic correlation has been found between gene repression and hypermethylation, several genes related to liver metabolism were either hypermethylated (Acsl4, Gpr40, Cel, Pparδ, Abca6, Ces3a, Sgms2) or hypomethylated (Soga1, Gpihbp1, Nr1d2, Mlxipl, Rps6kb2, Esrrb, Thra, Cidec). In specific cases (Hapln4, ApoA4, Cidec, genes involved in lipid metabolism and liver fibrosis) mRNA upregulation was associated with hypomethylation. In conclusion, we show for the first time wide disruptive physiological effects of low-dose of BPS, which raises the question of its harmlessness as an industrial substitute for BPA.
Collapse
Affiliation(s)
- Axelle Brulport
- Université de Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; AgroSup, LNC UMR1231, F-21000, Dijon, France; Nutrition Physiology and Toxicology Team (NUTox), INSERM, LNC UMR1231, F-21000, Dijon, France
| | - Daniel Vaiman
- From Gametes to Birth Team (FGTB), INSERM, U1016, Institut Cochin, F-75014, Paris, France; CNRS UMR8104, F-75014, Paris, France; Université Sorbonne Paris Cité, F-75014, Paris, France
| | - Marie-Christine Chagnon
- Université de Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; AgroSup, LNC UMR1231, F-21000, Dijon, France; Nutrition Physiology and Toxicology Team (NUTox), INSERM, LNC UMR1231, F-21000, Dijon, France
| | - Ludovic Le Corre
- Université de Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; AgroSup, LNC UMR1231, F-21000, Dijon, France; Nutrition Physiology and Toxicology Team (NUTox), INSERM, LNC UMR1231, F-21000, Dijon, France.
| |
Collapse
|
44
|
Herrera-Marcos LV, Sancho-Knapik S, Gabás-Rivera C, Barranquero C, Gascón S, Romanos E, Martínez-Beamonte R, Navarro MA, Surra JC, Arnal C, García-de-Jalón JA, Rodríguez-Yoldi MJ, Tena-Sempere M, Sánchez-Ramos C, Monsalve M, Osada J. Pgc1a is responsible for the sex differences in hepatic Cidec/Fsp27β mRNA expression in hepatic steatosis of mice fed a Western diet. Am J Physiol Endocrinol Metab 2020; 318:E249-E261. [PMID: 31846369 DOI: 10.1152/ajpendo.00199.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hepatic fat-specific protein 27 [cell death-inducing DNA fragmentation effector protein C (Cidec)/Fsp27] mRNA levels have been associated with hepatic lipid droplet extent under certain circumstances. To address its hepatic expression under different dietary conditions and in both sexes, apolipoprotein E (Apoe)-deficient mice were subjected to different experimental conditions for 11 wk to test the influence of cholesterol, Western diet, squalene, oleanolic acid, sex, and surgical castration on Cidec/Fsp27 mRNA expression. Dietary cholesterol increased hepatic Cidec/Fsp27β expression, an effect that was suppressed when cholesterol was combined with saturated fat as represented by Western diet feeding. Using the latter diet, neither oleanolic acid nor squalene modified its expression. Females showed lower levels of hepatic Cidec/Fsp27β expression than males when they were fed Western diets, a result that was translated into a lesser amount of CIDEC/FSP27 protein in lipid droplets and microsomes. This was also confirmed in low-density lipoprotein receptor (Ldlr)-deficient mice. Incubation with estradiol resulted in decreased Cidec/Fsp27β expression in AML12 cells. Whereas male surgical castration did not modify the expression, ovariectomized females did show increased levels compared with control females. Females also showed increased expression of peroxisome proliferator-activated receptor-γ coactivator 1-α (Pgc1a), suppressed by ovariectomy, and the values were significantly and inversely associated with those of Cidec/Fsp27β. When Pgc1a-deficient mice were used, the sex differences in Cidec/Fsp27β expression disappeared. Therefore, hepatic Cidec/Fsp27β expression has a complex regulation influenced by diet and sex hormonal milieu. The mRNA sex differences are controlled by Pgc1a.
Collapse
Affiliation(s)
- Luis V Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Sara Sancho-Knapik
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Clara Gabás-Rivera
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sonia Gascón
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Romanos
- Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María A Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquín C Surra
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Huesca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - José A García-de-Jalón
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - María J Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Tena-Sempere
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba e Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Sánchez-Ramos
- Instituto de Investigaciones Biomedicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - María Monsalve
- Instituto de Investigaciones Biomedicas "Alberto Sols," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, Centro de Investigación y Tecnología Agroalimentaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
45
|
Zhao J, Zhang X, Gao T, Wang S, Hou Y, Yuan P, Yang Y, Yang T, Xing J, Li J, Liu S. SIK2 enhances synthesis of fatty acid and cholesterol in ovarian cancer cells and tumor growth through PI3K/Akt signaling pathway. Cell Death Dis 2020; 11:25. [PMID: 31932581 PMCID: PMC6957524 DOI: 10.1038/s41419-019-2221-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022]
Abstract
Salt-inducible kinase 2 (SIK2) has been established as a regulator of diverse biological processes including cell metabolism. A recent study has reported that SIK2 is required for adipocyte-induced ovarian cancer (OC) survival through facilitating fatty acid oxidation. However, whether SIK2 also plays a role in the lipid synthesis in OC cells remains elusive. Here, we showed that SIK2 significantly promoted the lipid synthesis in OC cells. On the one hand, SIK2 enhanced fatty acid synthesis through upregulating the expression of sterol regulatory element binding protein 1c (SREBP1c) and thus the transcription of major lipogenic enzyme FASN. On the other hand, SIK2 promoted cholesterol synthesis through upregulating the expression of sterol regulatory element binding protein 2 (SREBP2) and thus the transcription of major cholesterol synthesis enzymes HMGCR. Moreover, PI3K/Akt signaling pathway was found to be involved in the upregulation of SREBP1c and SREBP2 in OC cells. Moreover, in vitro and in vivo assays indicated that the SIK2-regulated fatty acid and cholesterol synthesis played a critical role in the growth of OC cells. Our findings demonstrate that SIK2 is a critical regulator of lipid synthesis in OC cells and thus promotes OC growth, which provides a strong line of evidence for this molecule to be used as a therapeutic target in the treatment of this malignancy.
Collapse
Affiliation(s)
- Jing Zhao
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiaohong Zhang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Tian Gao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Shanci Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastorenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiran Hou
- Medical College of Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Peng Yuan
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Pain Treatment, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Yi Yang
- Department of Pain Treatment, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Tao Yang
- Department of Pain Treatment, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jibin Li
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Shujuan Liu
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
46
|
Wu J, Frazier K, Zhang J, Gan Z, Wang T, Zhong X. Emerging role of m 6 A RNA methylation in nutritional physiology and metabolism. Obes Rev 2020; 21:e12942. [PMID: 31475777 PMCID: PMC7427634 DOI: 10.1111/obr.12942] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022]
Abstract
N6 -methyladenine (m6 A) is the most prevalent type of internal RNA methylation in eukaryotic mRNA and plays critical roles in regulating gene expression for fundamental cellular processes and diverse physiological functions. Recent evidence indicates that m6 A methylation regulates physiology and metabolism, and m6 A has been increasingly implicated in a variety of human diseases, including obesity, diabetes, metabolic syndrome and cancer. Conversely, nutrition and diet can modulate or reverse m6 A methylation patterns on gene expression. In this review, we summarize the recent progress in the study of the m6 A methylation mechanisms and highlight the crosstalk between m6 A modification, nutritional physiology and metabolism.
Collapse
Affiliation(s)
- Jiamin Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu, Nanjing, 210095, PR China
| | - Katya Frazier
- Department of Medicine, University of Chicago. Chicago, IL 60637, USA
| | - Jingfei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu, Nanjing, 210095, PR China
| | - Zhending Gan
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu, Nanjing, 210095, PR China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu, Nanjing, 210095, PR China
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu, Nanjing, 210095, PR China
| |
Collapse
|
47
|
Petito-da-Silva TI, Souza-Mello V, Barbosa-da-Silva S. Empaglifozin mitigates NAFLD in high-fat-fed mice by alleviating insulin resistance, lipogenesis and ER stress. Mol Cell Endocrinol 2019; 498:110539. [PMID: 31419466 DOI: 10.1016/j.mce.2019.110539] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/09/2019] [Accepted: 08/11/2019] [Indexed: 12/14/2022]
Abstract
AIM To evaluate the pleiotropic effects of empagliflozin in the liver through lipogenesis, beta-oxidation, and endoplasmic reticulum stress pathways. METHODS Male C57Bl/6 mice, 3 months of age, received a control diet (C, 10% lipids, n = 20) or high-fat diet (HF, 50% lipids, n = 20) for 10 weeks, after that, the groups were subdivided to receive empagliflozin, during 5 weeks at a dose of 10 mg/kg/day added to the diets, totalizing four groups: C, C-EMPA, HF, and HF-EMPA. We performed biochemical analyzes, oral glucose tolerance test, homeostasis model assessment of insulin resistance (HOMA-IR), indirect calorimetry, liver stereology, western blotting, RT-qPCR for genes related to beta-oxidation, lipogenesis, and endoplasmic reticulum stress. RESULTS After the treatment with empagliflozin, there was a 4% increase in energy expenditure, a 5% reduction in body mass, improvement in glucose tolerance and insulin sensitivity and insulin resistance. The expression of Ppar alpha was greater in the HF-EMPA group with a concomitant reduction in the expression of the lipogenic genes Fas, Srebp1c and Ppar gamma, according to protein expression. In addition, HF-EMPA showed a reduction in the genes related to endoplasmic reticulum stress Chop, Atf4, and Gadd45. CONCLUSION Empagliflozin mitigates the development of NAFLD, confirmed through reduced expression of the genes involved in hepatic lipogenesis and genes involved in endoplasmic reticulum stress. Thus, empagliflozin may be an important tool to treat the progression of hepatic steatosis.
Collapse
Affiliation(s)
- Tamiris Ingrid Petito-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, State University of Rio de Janeiro, RJ, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, State University of Rio de Janeiro, RJ, Brazil
| | - Sandra Barbosa-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, State University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
48
|
Liu YT, Lai YH, Lin HH, Chen JH. Lotus Seedpod Extracts Reduced Lipid Accumulation and Lipotoxicity in Hepatocytes. Nutrients 2019; 11:E2895. [PMID: 31795130 PMCID: PMC6950491 DOI: 10.3390/nu11122895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely associated with metabolic disorders, including hepatic lipid accumulation and lipotoxicity. Plant-derived polyphenols have attracted considerable attention in the prevention of NAFLD. Lotus seedpod, rich in polyphenols, is a traditional Chinese herbal medicine. Previous studies have showed that lotus seedpod possess radioprotective, antioxidant, anti-cancer, and anti-inflammatory activities. In this study, the in vitro hepatoprotective effect of lotus seedpod extract (LSE) and its main component epigallocatechin (EGC) was examined. Firstly, oleic acid (OA), an unsaturated fatty acid, was used to induce the phenotype of NAFLD in human hepatocytes, HepG2 cells. LSE dose-dependently improved the OA-induced viability loss of HepG2 cells. Non-cytotoxic concentrations of LSE or EGC abolished intracellular lipid accumulation and oxidative stress in the OA-treated cells. In addition, LSE and EGC showed a minor effect on autophagy, and potential in reducing OA-induced occurrence of apoptosis confirmed by morphological and biochemical features, including an increase in the formation of apoptotic bodies, the exposure of phosphatidylserine, and activation of caspases. Molecular data showed the anti-apoptotic effect of LSE might be mediated via downregulation of the mitochondrial pathway. Our data imply that EGC-enriched LSE potentially could be developed as an anti-NAFLD agent.
Collapse
Affiliation(s)
- Yen-Tze Liu
- Department of Family Medicine, Changhua Christian Hospital, No. 135 Nanhsiao Street, Changhua City 50006, Taiwan;
| | - Yen-Hsun Lai
- Department of Nutrition, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung City 40201, Taiwan;
| | - Hui-Hsuan Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung City 40201, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung City 40201, Taiwan
| | - Jing-Hsien Chen
- Department of Nutrition, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung City 40201, Taiwan;
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung City 40201, Taiwan
| |
Collapse
|
49
|
Zhou L, Li Q, Chen A, Liu N, Chen N, Chen X, Zhu L, Xia B, Gong Y, Chen X. KLF15-activating Twist2 ameliorated hepatic steatosis by inhibiting inflammation and improving mitochondrial dysfunction via NF-κB-FGF21 or SREBP1c-FGF21 pathway. FASEB J 2019; 33:14254-14269. [PMID: 31648561 DOI: 10.1096/fj.201901347rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Twist-related protein 2 (TWIST2) is identified as a basic helix-loop-helix (b-HLH) transcription repressor by dimerizing with other b-HLH proteins. The significance of TWIST2 has been emphasized in various tumors; however, few studies report its functions in metabolism and metabolic diseases. Here we aimed to explore the novel role and regulation mechanism of TWIST2 in hepatic steatosis. Our results showed that Twist2 knockdown caused mice obesity, insulin resistance, and hepatic steatosis, which were accompanied with inflammation, endoplasmic reticulum stress, and mitochondrial dysfunction. In vitro, TWIST2 overexpression ameliorated hepatocellular steatosis, inhibited inflammation, and improved mitochondrial content and function with a fibroblast growth factor 21 (FGF21)-dependent pattern. NF-κB negatively regulated FGF21 transcription by directly binding to FGF21 promoter DNA, which was eliminated by TWIST2 overexpression by inhibiting NF-κB expression and translocation to nucleus. TWIST2 overexpression decreased intracellular reactive oxygen species level, increased mitochondrial DNA and biogenesis, and enhanced ATP production and antioxidation ability. Additionally, TWIST2 expression was repressed by insulin-targeting sterol regulatory element-binding protein 1c (SREBP1c) and forkhead box protein O1 and was enhanced by dexamethasone targeting Krüppel-like factor 15, which directly interacted with Twist2 promoter DNA. Together, our studies identify an important role and regulation mechanism of TWIST2 in maintaining hepatic homeostasis by ameliorating steatosis, inflammation, and oxidative stress via the NF-κB-FGF21 or SREBP1c-FGF21 pathway, which may provide a new therapeutic scheme for nonalcoholic fatty liver disease.-Zhou, L., Li, Q., Chen, A., Liu, N., Chen, N., Chen, X., Zhu, L., Xia, B., Gong, Y., Chen, X. KLF15-activating Twist2 ameliorated hepatic steatosis by inhibiting inflammation and improving mitochondrial dysfunction via NF-κB-FGF21 or SREBP1c-FGF21 pathway.
Collapse
Affiliation(s)
- Lulu Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Qinjin Li
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ao Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Na Liu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ning Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaojun Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Lin Zhu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Benzeng Xia
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yuqing Gong
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction, Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
50
|
Zhang J, Zhang SD, Wang P, Guo N, Wang W, Yao LP, Yang Q, Efferth T, Jiao J, Fu YJ. Pinolenic acid ameliorates oleic acid-induced lipogenesis and oxidative stress via AMPK/SIRT1 signaling pathway in HepG2 cells. Eur J Pharmacol 2019; 861:172618. [DOI: 10.1016/j.ejphar.2019.172618] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/20/2022]
|