1
|
Bamahel AS, Sun X, Wu W, Mu C, Liu J, Bi S, Xu H. Regulatory Roles and Therapeutic Potential of miR-122-5p in Hypoxic-Ischemic Brain Injury: Comprehensive Review. Cell Biochem Biophys 2025:10.1007/s12013-025-01686-6. [PMID: 40016565 DOI: 10.1007/s12013-025-01686-6] [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] [Accepted: 01/27/2025] [Indexed: 03/01/2025]
Abstract
In the regulation of gene expression, epigenetic factors, including non-coding RNAs (ncRNAs) play a role in genetics. Among the ncRNA family, microRNAs (miRNAs) have gained significant attention for their involvement in post-transcriptional gene regulation, with profound implications for both normal and pathological processes including neurological diseases such as hypoxic-ischemic brain injury. A specific miRNA, called miR-122-5p, has gained attention in hypoxic-ischemic conditions, where it modulates critical pathways such as inflammation, oxidative stress, and neuronal survival. The purpose of this review is to highlight recent advances in the biogenesis, expression, and regulation of miR-122-5p, focusing on its role in hypoxic-ischemic conditions and its potential as a therapeutic target. We first studied the therapeutic strategies and potential clinical applications of miR-122-5p, our research showing it interacts with key transcription factors, such as HIF-1α and NF-κB, influencing cellular responses to low oxygen levels. Our findings revealed that miR-122-5p plays a vital role in hypoxic-ischemic brain injury, with its abnormal levels strongly associated with increased brain damage and neuroinflammation, suggesting its potential as a promising therapeutic target. Furthermore, miR-122-5p influences various biological processes in the brain, such as metabolism and blood vessel formation. The use of miR-122-5p inhibitor has been shown to increase autophagy, reduce apoptosis, and decrease oxidative stress and inflammation, thereby protecting neurons and improving outcomes in hypoxic encephalopathy by targeting multiple genes related to these processes. Conversely, miR-122-5p mimics exacerbate oxidative stress and reduce autophagy. These findings highlight the therapeutic potential of miR-122-5p inhibition in reducing brain injury and promoting recovery in hypoxic-ischemic encephalopathy through enhanced neuroprotective mechanisms and the suppression of harmful cellular processes. However, further experimental studies are needed to fully understand the therapeutic potential of targeting miR-122-5p and its related genes in hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
| | - Xun Sun
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Wei Wu
- Public Health College, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Chenxi Mu
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Jia Liu
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Sheng Bi
- Clinical Medical College, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Hui Xu
- Basic Medical College, Jiamusi University, Jiamusi, Heilongjiang, China.
| |
Collapse
|
2
|
Lu L, Hua W, Li F, Che Z, Tian M, Lu YY, Chi Q, Zhang J, Huang Q. Arsenic Exposure Triggers Nonalcoholic Fatty Liver Disease through Repressing S-Adenosylmethionine-Dependent Histone Methylation in Rats. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:268-278. [PMID: 39746780 DOI: 10.1021/acs.est.4c10417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Arsenic (As) is a toxic metalloid widespread in the environment, and its exposure has been associated with a variety of adverse health outcomes. As exposure is demonstrated to cause nonalcoholic fatty liver disease (NAFLD), and the underlying epigenetic mechanisms remain largely unknown. This study aimed to investigate the roles of histone modifications in low-level As exposure-induced NAFLD in rats. The results showed that exposure to As caused lipid accumulation and upregulated the expression of lipid metabolism-related genes Fabp1, Srebf1, and Apoc3, while downregulated Acox1 and Cpt1a in rat liver. In addition, it was found that inorganic arsenite (iAsIII) was methylated to DMA, and the S-adenosylmethionine (SAM) level was decreased, which would contribute to the repression of H3K9me1/2 in rat liver after exposure. The in vitro studies revealed that SAM supplementation attenuated lipid accumulation by restoring H3K9me1/2 in HepG2 cells, which further confirmed our animal results. Therefore, it is suggested that As methylation depleted SAM, which inhibited H3K9me1/2 and activated Fabp1, Srebf1, and Apoc3 expressions, leading to NAFLD upon inorganic As exposure. Overall, these data shed new light on the role of SAM-mediated histone methylation in As-triggered NAFLD, which could be useful for the prevention and intervention of hepatotoxicity induced by environmental As exposure.
Collapse
Affiliation(s)
- Lu Lu
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Weizhen Hua
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Fuping Li
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Zhenbin Che
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Meiping Tian
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yan-Yang Lu
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Qiaoqiao Chi
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Jie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qingyu Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| |
Collapse
|
3
|
Dong QQ, Yang Y, Tao H, Lu C, Yang JJ. m6A epitranscriptomic and epigenetic crosstalk in liver fibrosis: Special emphasis on DNA methylation and non-coding RNAs. Cell Signal 2024; 122:111302. [PMID: 39025344 DOI: 10.1016/j.cellsig.2024.111302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Liver fibrosis is a pathological process caused by a variety of chronic liver diseases. Currently, therapeutic options for liver fibrosis are very limited, highlighting the urgent need to explore new treatment approaches. Epigenetic modifications and epitranscriptomic modifications, as reversible regulatory mechanisms, are involved in the development of liver fibrosis. In recent years, researches in epitranscriptomics and epigenetics have opened new perspectives for understanding the pathogenesis of liver fibrosis. Exploring the epigenetic mechanisms of liver fibrosis may provide valuable insights into the development of new therapies for chronic liver diseases. This review primarily focus on the regulatory mechanisms of N6-methyladenosine (m6A) modification, non-coding RNA, and DNA methylation in organ fibrosis. It discusses the interactions between m6A modification and DNA methylation, as well as between m6A modification and non-coding RNA, providing a reference for understanding the interplay between epitranscriptomics and epigenetics.
Collapse
Affiliation(s)
- Qi-Qi Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yang Yang
- Department of General Surgery, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, China
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan 232001, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
4
|
Kim JY, Yang AY, Kim K, Kwon HH, Leem J, Kim YA. Pharmacological inhibition of p300 ameliorates steatosis, inflammation, and fibrosis in mice with non-alcoholic steatohepatitis. Heliyon 2024; 10:e30908. [PMID: 38774067 PMCID: PMC11107220 DOI: 10.1016/j.heliyon.2024.e30908] [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: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
The histone acetyltransferase p300 plays a pivotal role in regulating gene expression and cellular phenotype through epigenetic mechanisms. It significantly influences lipid metabolism, which is a key factor in the pathogenesis of non-alcoholic steatohepatitis (NASH), by modulating the transcription of genes involved in lipid synthesis and accumulation. This study aimed to investigate the protective potential of inhibiting p300 in NASH. Male C57BL/6J mice were subjected to a methionine- and choline-deficient (MCD) diet for 4 weeks to induce NASH, and during this period, the p300 inhibitor C646 (10 mg/kg) was administered three times a week. C646 treatment reduced the elevation of p300 expression and histone H3 acetylation, leading to a decrease in liver injury markers in the serum and an improvement in the histological abnormalities observed in MCD diet-fed mice. C646 also reduced lipid accumulation by modulating de novo lipogenesis and suppressed inflammation, including cytokine overproduction and macrophage infiltration. Furthermore, C646 mitigated liver fibrosis and myofibroblast accumulation. This protective effect was achieved through the inhibition of apoptosis by reducing p53 and Bax expression and the suppression of ferroptosis by decreasing lipid peroxidation while enhancing antioxidant defenses. Additionally, C646 alleviated endoplasmic reticulum stress, as evidenced by the downregulation of unfolded protein response signaling molecules. These results highlight the potential of p300 as a therapeutic target for NASH.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Ah Young Yang
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Kiryeong Kim
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Hyun Hee Kwon
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Yun-A Kim
- Department of Family Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| |
Collapse
|
5
|
Nikolic A, Fahlbusch P, Riffelmann NK, Wahlers N, Jacob S, Hartwig S, Kettel U, Schiller M, Dille M, Al-Hasani H, Kotzka J, Knebel B. Chronic stress alters hepatic metabolism and thermodynamic respiratory efficiency affecting epigenetics in C57BL/6 mice. iScience 2024; 27:109276. [PMID: 38450153 PMCID: PMC10915629 DOI: 10.1016/j.isci.2024.109276] [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: 08/18/2023] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 03/08/2024] Open
Abstract
Chronic stress episodes increase metabolic disease risk even after recovery. We propose that persistent stress detrimentally impacts hepatic metabolic reprogramming, particularly mitochondrial function. In male C57BL/6 mice chronic variable stress (Cvs) reduced energy expenditure (EE) and body mass despite increased energy intake versus controls. This coincided with decreased glucose metabolism and increased lipid β-oxidation, correlating with EE. After Cvs, mitochondrial function revealed increased thermodynamic efficiency (ƞ-opt) of complex CI, positively correlating with blood glucose and NEFA and inversely with EE. After Cvs recovery, the metabolic flexibility of hepatocytes was lost. Reduced CI-driving NAD+/NADH ratio, and diminished methylation-related one-carbon cycle components hinted at epigenetic regulation. Although initial DNA methylation differences were minimal after Cvs, they diverged during the recovery phase. Here, the altered enrichment of mitochondrial DNA methylation and linked transcriptional networks were observed. In conclusion, Cvs rapidly initiates the reprogramming of hepatic energy metabolism, supported by lasting epigenetic modifications.
Collapse
Affiliation(s)
- Aleksandra Nikolic
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Pia Fahlbusch
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Nele-Kathrien Riffelmann
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
| | - Natalie Wahlers
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
| | - Sylvia Jacob
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
| | - Sonja Hartwig
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Ulrike Kettel
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
| | - Martina Schiller
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
| | - Matthias Dille
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
| | - Hadi Al-Hasani
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
- Medical Faculty Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jörg Kotzka
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Birgit Knebel
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| |
Collapse
|
6
|
Zhao P, Yang W, Xiao H, Zhang S, Gao C, Piao H, Liu L, Li S. Vitamin K2 protects mice against non-alcoholic fatty liver disease induced by high-fat diet. Sci Rep 2024; 14:3075. [PMID: 38321064 PMCID: PMC10847165 DOI: 10.1038/s41598-024-53644-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/03/2024] [Indexed: 02/08/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide and there is a huge unmet need to find safer and more effective drugs. Vitamin K has been found to regulate lipid metabolism in the liver. However, the effects of vitamin K2 on NAFLD is unclear. This study aims to evaluate the preventive and therapeutic effects of vitamin K2 in the process of fatty liver formation and to explore molecular mechanisms the associated with lipid metabolism. A non-alcoholic fatty liver model was established by high-fat diet administration for three months. Vitamin K2 significantly reduced the body weight, abdominal circumference and body fat percentage of NAFLD mice. Vitamin K2 also showed histological benefits in reducing hepatic steatosis. NAFLD mice induced by high-fat diet showed increased HMGR while vitamin K2 intervention could reverse the pathological lterations. Adiponectin (APN) is an endogenous bioactive polypeptide or protein secreted by adipocytes. We detected APN, SOD, AlaDH and other indicators that may affect the state of high-fat diet mice, but the experimental results showed that the above indicators did not change significantly. It is worth noting that the effect of vitamin K2 supplementation on the lipid-lowering effect of uc OC in vivo needs to be further explored. This study first reported the protective effect of vitamin K2 on high-fat diet-induced NAFLD in mice. The protective effect of vitamin K2 may be related to the improvement of lipid metabolism disorder in NAFLD.
Collapse
Affiliation(s)
- Peizuo Zhao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Weidong Yang
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Huiyu Xiao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Shuaishuai Zhang
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Chuanzhou Gao
- Central Laboratory, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Hua Piao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Lihong Liu
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, People's Republic of China.
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, People's Republic of China.
| |
Collapse
|
7
|
Caputo V, Tarantino G, Santini SJ, Fracassi G, Balsano C. The Role of Epigenetic Control of Mitochondrial (Dys)Function in MASLD Onset and Progression. Nutrients 2023; 15:4757. [PMID: 38004151 PMCID: PMC10675587 DOI: 10.3390/nu15224757] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/06/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Metabolic dysfunction-associated steatotic fatty liver disease (MASLD), a novel definition for NAFLD, represents one of the most common causes of liver disease, and its incidence is increasing worldwide. It is characterized by a complex etiopathogenesis in which mitochondrial dysfunction exerts a pivotal role together with alteration of lipid metabolism, inflammation, and oxidative stress. Nutrients and bioactive compounds can influence such mechanisms so that changes in diet and lifestyle are regarded as important treatment strategies. Notably, natural compounds can exert their influence through changes of the epigenetic landscape, overall resulting in rewiring of molecular networks involved in cell and tissue homeostasis. Considering such information, the present review aims at providing evidence of epigenetic modifications occurring at mitochondria in response to natural and bioactive compounds in the context of liver (dys)function. For this purpose, recent studies reporting effects of compounds on mitochondria in the context of NAFLD/MASLD, as well as research showing alteration of DNA methylation and non-coding RNAs-related circuits occurring at liver mitochondria, will be illustrated. Overall, the present review will highlight the importance of understanding the bioactive compounds-dependent epigenetic modulation of mitochondria for improving the knowledge of MASLD and identifying biomarkers to be employed for effective preventative strategies or treatment protocols.
Collapse
Affiliation(s)
- Valerio Caputo
- Department of Life, Health and Environmental Sciences-MESVA, School of Emergency-Urgency Medicine, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (S.J.S.); (G.F.)
- F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy
| | - Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University of Naples, 80138 Naples, Italy;
| | - Silvano Junior Santini
- Department of Life, Health and Environmental Sciences-MESVA, School of Emergency-Urgency Medicine, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (S.J.S.); (G.F.)
- F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy
| | - Giovanna Fracassi
- Department of Life, Health and Environmental Sciences-MESVA, School of Emergency-Urgency Medicine, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (S.J.S.); (G.F.)
| | - Clara Balsano
- Department of Life, Health and Environmental Sciences-MESVA, School of Emergency-Urgency Medicine, University of L’Aquila, 67100 L’Aquila, Italy; (V.C.); (S.J.S.); (G.F.)
- F. Balsano Foundation, Via Giovanni Battista Martini 6, 00198 Rome, Italy
| |
Collapse
|
8
|
Zhang T, Nie Y, Wang J. The emerging significance of mitochondrial targeted strategies in NAFLD treatment. Life Sci 2023; 329:121943. [PMID: 37454757 DOI: 10.1016/j.lfs.2023.121943] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease worldwide, ranging from liver steatosis to nonalcoholic steatohepatitis, which ultimately progresses to fibrosis, cirrhosis, and hepatocellular carcinoma. Individuals with NAFLD have a higher risk of developing cardiovascular and extrahepatic cancers. Despite the great progress being made in understanding the pathogenesis and the introduction of new pharmacological targets for NAFLD, no drug or intervention has been accepted for its management. Recent evidence suggests that NAFLD may be a mitochondrial disease, as mitochondrial dysfunction is involved in the pathological processes that lead to NAFLD. In this review, we describe the recent advances in our understanding of the mechanisms associated with mitochondrial dysfunction in NAFLD progression. Moreover, we discuss recent advances in the efficacy of mitochondria-targeted compounds (e.g., Mito-Q, MitoVit-E, MitoTEMPO, SS-31, mitochondrial uncouplers, and mitochondrial pyruvate carrier inhibitors) for treating NAFLD. Furthermore, we present some medications currently being tested in clinical trials for NAFLD treatment, such as exercise, mesenchymal stem cells, bile acids and their analogs, and antidiabetic drugs, with a focus on their efficacy in improving mitochondrial function. Based on this evidence, further investigations into the development of mitochondria-based agents may provide new and promising alternatives for NAFLD management.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yingli Nie
- Department of Dermatology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China.
| | - Jiliang Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
9
|
Aggeletopoulou I, Kalafateli M, Tsounis EP, Triantos C. Epigenetic Regulation in Lean Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:12864. [PMID: 37629043 PMCID: PMC10454848 DOI: 10.3390/ijms241612864] [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: 07/31/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most prominent cause of chronic liver disease worldwide, is a rapidly growing epidemic. It consists of a wide range of liver diseases, from steatosis to nonalcoholic steatohepatitis, and predisposes patients to liver fibrosis, cirrhosis, and even hepatocellular carcinoma. NAFLD is strongly correlated with obesity; however, it has been extensively reported among lean/nonobese individuals in recent years. Although lean patients demonstrate a lower prevalence of diabetes mellitus, central obesity, dyslipidemia, hypertension, and metabolic syndrome, a percentage of these patients may develop steatohepatitis, advanced liver fibrosis, and cardiovascular disease, and have increased all-cause mortality. The pathophysiological mechanisms of lean NAFLD remain vague. Studies have reported that lean NAFLD demonstrates a close association with environmental factors, genetic predisposition, and epigenetic modifications. In this review, we aim to discuss and summarize the epigenetic mechanisms involved in lean NAFLD and to introduce the interaction between epigenetic patterns and genetic or non genetic factors. Several epigenetic mechanisms have been implicated in the regulation of lean NAFLD. These include DNA methylation, histone modifications, and noncoding-RNA-mediated gene regulation. Epigenetics is an area of special interest in the setting of lean NAFLD as it could provide new insights into the therapeutic options and noninvasive biomarkers that target this under-recognized and challenging disorder.
Collapse
Affiliation(s)
- Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (E.P.T.)
| | - Maria Kalafateli
- Department of Gastroenterology, General Hospital of Patras, 26332 Patras, Greece;
| | - Efthymios P. Tsounis
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (E.P.T.)
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (I.A.); (E.P.T.)
| |
Collapse
|
10
|
Zheng Y, Wang S, Wu J, Wang Y. Mitochondrial metabolic dysfunction and non-alcoholic fatty liver disease: new insights from pathogenic mechanisms to clinically targeted therapy. J Transl Med 2023; 21:510. [PMID: 37507803 PMCID: PMC10375703 DOI: 10.1186/s12967-023-04367-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is among the most widespread metabolic disease globally, and its associated complications including insulin resistance and diabetes have become threatening conditions for human health. Previous studies on non-alcoholic fatty liver disease (NAFLD) were focused on the liver's lipid metabolism. However, growing evidence suggests that mitochondrial metabolism is involved in the pathogenesis of NAFLD to varying degrees in several ways, for instance in cellular division, oxidative stress, autophagy, and mitochondrial quality control. Ultimately, liver function gradually declines as a result of mitochondrial dysfunction. The liver is unable to transfer the excess lipid droplets outside the liver. Therefore, how to regulate hepatic mitochondrial function to treat NAFLD has become the focus of current research. This review provides details about the intrinsic link of NAFLD with mitochondrial metabolism and the mechanisms by which mitochondrial dysfunctions contribute to NAFLD progression. Given the crucial role of mitochondrial metabolism in NAFLD progression, the application potential of multiple mitochondrial function improvement modalities (including physical exercise, diabetic medications, small molecule agonists targeting Sirt3, and mitochondria-specific antioxidants) in the treatment of NAFLD was evaluated hoping to provide new insights into NAFLD treatment.
Collapse
Affiliation(s)
- Youwei Zheng
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Shiting Wang
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jialiang Wu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
11
|
Lei P, Hu N, Wu Y, Tang M, Lin C, Kong L, Zhang L, Luo P, Chan LW. Radiobioinformatics: A novel bridge between basic research and clinical practice for clinical decision support in diffuse liver diseases. IRADIOLOGY 2023; 1:167-189. [DOI: 10.1002/ird3.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/18/2023] [Indexed: 01/04/2025]
Abstract
AbstractThe liver is a multifaceted organ that is responsible for many critical functions encompassing amino acid, carbohydrate, and lipid metabolism, all of which make a healthy liver essential for the human body. Contemporary imaging methodologies have remarkable diagnostic accuracy in discerning focal liver lesions; however, a comprehensive understanding of diffuse liver diseases is a requisite for radiologists to accurately diagnose or predict the progression of such lesions within clinical contexts. Nonetheless, the conventional attributes of radiological features, including morphology, size, margin, density, signal intensity, and echoes, limit their clinical utility. Radiomics is a widely used approach that is characterized by the extraction of copious image features from radiographic depictions, which gives it considerable potential in addressing this limitation. It is worth noting that functional or molecular alterations occur significantly prior to the morphological shifts discernible by imaging modalities. Consequently, the explication of potential mechanisms by multiomics analyses (encompassing genomics, epigenomics, transcriptomics, proteomics, and metabolomics) is essential for investigating putative signal pathway regulations from a radiological viewpoint. In this review, we elaborate on the principal pathological categorizations of diffuse liver diseases, the evaluation of multiomics approaches pertaining to diffuse liver diseases, and the prospective value of predictive models. Accordingly, the overarching objective of this review is to scrutinize the interrelations between radiological features and bioinformatics as well as to consider the development of prediction models predicated on radiobioinformatics as integral components of clinical decision support systems for diffuse liver diseases.
Collapse
Affiliation(s)
- Pinggui Lei
- Department of Health Technology and Informatics The Hong Kong Polytechnic University Kowloon Hong Kong SAR China
- Department of Radiology The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
- School of Public Health Guizhou Medical University Guiyang Guizhou China
| | - Na Hu
- Department of Radiology The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Yuhui Wu
- Department of Radiology The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Maowen Tang
- Department of Radiology The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Chong Lin
- Department of Radiology The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Luoyi Kong
- Department of Health Technology and Informatics The Hong Kong Polytechnic University Kowloon Hong Kong SAR China
| | - Lingfeng Zhang
- Department of Health Technology and Informatics The Hong Kong Polytechnic University Kowloon Hong Kong SAR China
| | - Peng Luo
- School of Public Health Guizhou Medical University Guiyang Guizhou China
| | - Lawrence Wing‐Chi Chan
- Department of Health Technology and Informatics The Hong Kong Polytechnic University Kowloon Hong Kong SAR China
| |
Collapse
|