1
|
Raju C, Sankaranarayanan K. Insights on post-translational modifications in fatty liver and fibrosis progression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167659. [PMID: 39788217 DOI: 10.1016/j.bbadis.2025.167659] [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: 09/06/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease [MASLD] is a pervasive multifactorial health burden. Post-translational modifications [PTMs] of amino acid residues in protein domains demonstrate pivotal roles for imparting dynamic alterations in the cellular micro milieu. The crux of identifying novel druggable targets relies on comprehensively studying the etiology of metabolic disorders. This review article presents how different chemical moieties of various PTMs like phosphorylation, methylation, ubiquitination, glutathionylation, neddylation, acetylation, SUMOylation, lactylation, crotonylation, hydroxylation, glycosylation, citrullination, S-sulfhydration and succinylation presents the cause-effect contribution towards the MASLD spectra. Additionally, the therapeutic prospects in the management of liver steatosis and hepatic fibrosis via targeting PTMs and regulatory enzymes are also encapsulated. This review seeks to understand the function of protein modifications in progression and promote the markers discovery of diagnostic, prognostic and drug targets towards MASLD management which could also halt the progression of a catalogue of related diseases.
Collapse
Affiliation(s)
- Chithra Raju
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India
| | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India.
| |
Collapse
|
2
|
Su Y, Hou C, Wang M, Ren K, Zhou D, Liu X, Zhao S, Liu X. Metformin induces mitochondrial fission and reduces energy metabolism by targeting respiratory chain complex I in hepatic stellate cells to reverse liver fibrosis. Int J Biochem Cell Biol 2023; 157:106375. [PMID: 36716817 DOI: 10.1016/j.biocel.2023.106375] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/27/2022] [Accepted: 01/26/2023] [Indexed: 01/30/2023]
Abstract
The activation and proliferation of hepatic stellate cells (HSCs) are critical processes for the treatment of liver fibrosis. It is necessary to identify effective drugs for the treatment of liver fibrosis and elucidate their mechanisms of action. Metformin can inhibit HSCs; however, no systematic studies demonstrating the effects of metformin on mitochondria in HSCs have been reported. This study demonstrated that metformin induces mitochondrial fission by phosphorylating AMPK/DRP1 (S616) in HSCs to decrease the expression of α-SMA and collagen. Additionally, metformin repressed the total ATP production rate, especially the production rate of ATP produced through mitochondrial oxidative phosphorylation, by inhibiting the enzymatic activity of complex I. Further analysis revealed that metformin strongly constrained the transcription of mitochondrial genes (ND1-ND6 and ND4L) that encode the core subunits of respiratory chain I. Upregulation of the mRNA expression of HK2 and GLUT1 slightly enhanced glycolysis. Additionally, metformin increased mitochondrial DNA (mtDNA) copy number to suppress the proliferation and activation of HSCs, indicating that mtDNA copy number can alter the fate of HSCs. In conclusion, metformin can induce mitochondrial fragmentation and low-level energy metabolism in HSCs, thereby suppressing HSCs activation and proliferation to reverse liver fibrosis.
Collapse
Affiliation(s)
- Ying Su
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chenjian Hou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meili Wang
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Kehan Ren
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Danmei Zhou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoli Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shanyu Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiuping Liu
- Department of Pathology, Shanghai Fifth People's Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200240, China.
| |
Collapse
|
3
|
Wan G, Chen Z, Lei L, Geng X, Zhang Y, Yang C, Cao W, Pan Z. The total polyphenolic glycoside extract of Lamiophlomis rotata ameliorates hepatic fibrosis through apoptosis by TGF-β/Smad signaling pathway. Chin Med 2023; 18:20. [PMID: 36829153 PMCID: PMC9951520 DOI: 10.1186/s13020-023-00723-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 02/07/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Hepatic fibrosis is characterized by the excessive deposition of extracellular matrix (ECM) which is mainly secreted by activated hepatic stellate cells (HSCs). Lamiophlomis rotata (L. rotata) was recorded to treat jaundice in the traditional Tibetan medical system with the potential of hepatoprotection. However, the bioactivities and the possible mechanism of L. rotata on hepatic fibrosis is still largely unknown. AIM OF THE STUDY To investigate the anti-hepatic fibrosis effects of bioactivities in L. rotata and the probable mechanism of action. MATERIALS AND METHODS Herein, total polyphenolic glycosides of L. rotata (TPLR) was purified with the selectivity adsorption resin and was analyzed by ultrahigh-performance liquid chromatography coupled with time-of-flight mass spectrometry (UPLC-Q/TOF/MSn). The anti-hepatic fibrosis effect of TPLR was evaluated by carbon tetrachloride (CCl4)-induced liver fibrosis, and was evaluated with the apoptosis of activated HSCs. RESULTS In total, sixteen compounds, including nine phenylpropanoids and six flavonoids, were identified in the UPLC-TOF-MSn profile of the extracts. TPLR significantly ameliorated hepatic fibrosis in CCl4-induced mice and inhibited HSCs proliferation, Moreover, TPLR notably increased the apoptosis of activated HSCs along with up-regulated caspase-3, -8, -9, and -10. Furthermore, TPLR inhibited TGF-β/Smad pathway ameliorating hepatic fibrosis though downregulation the expression of Smad2/3, Smad4, and upregulation the expression of Smad7 in vivo and in vitro. Simultaneously, the expression of fibronectin (FN), α-smooth muscle actin (α-SMA), and Collagen I (Col1α1) were decreased in tissues and in cells with TPLR administration. CONCLUSION These results initially demonstrated that TPLR has the potential to ameliorate hepatic fibrosis through an apoptosis mechanism via TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Guoguo Wan
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 People’s Republic of China
| | - Zhiwei Chen
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 People’s Republic of China
| | - Lei Lei
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 People’s Republic of China
| | - Xiaoyu Geng
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 People’s Republic of China
| | - Yi Zhang
- grid.411304.30000 0001 0376 205XCentre for Academic Inheritance and Innovation of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130 China
| | - Congwen Yang
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 People’s Republic of China
| | - Wenfu Cao
- grid.203458.80000 0000 8653 0555Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016 People’s Republic of China
| | - Zheng Pan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, No.1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
4
|
Tornesello ML, Tornesello AL, Starita N, Cerasuolo A, Izzo F, Buonaguro L, Buonaguro FM. Telomerase: a good target in hepatocellular carcinoma? An overview of relevant preclinical data. Expert Opin Ther Targets 2022; 26:767-780. [PMID: 36369706 DOI: 10.1080/14728222.2022.2147062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Napoli, Italy
| | - Anna Lucia Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Napoli, Italy
| | - Noemy Starita
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Napoli, Italy
| | - Andrea Cerasuolo
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Napoli, Italy
| | - Francesco Izzo
- Hepatobiliary Surgical Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, 80131 Naples, Italy
| | - Luigi Buonaguro
- Laboratory of Cancer Immunoregulation, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Napoli, Italy
| | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Napoli, Italy
| |
Collapse
|
5
|
Wang L, Feng J, Deng Y, Yang Q, Wei Q, Ye D, Rong X, Guo J. CCAAT/Enhancer-Binding Proteins in Fibrosis: Complex Roles Beyond Conventional Understanding. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9891689. [PMID: 36299447 PMCID: PMC9575473 DOI: 10.34133/2022/9891689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/18/2022] [Indexed: 07/29/2023]
Abstract
CCAAT/enhancer-binding proteins (C/EBPs) are a family of at least six identified transcription factors that contain a highly conserved basic leucine zipper domain and interact selectively with duplex DNA to regulate target gene expression. C/EBPs play important roles in various physiological processes, and their abnormal function can lead to various diseases. Recently, accumulating evidence has demonstrated that aberrant C/EBP expression or activity is closely associated with the onset and progression of fibrosis in several organs and tissues. During fibrosis, various C/EBPs can exert distinct functions in the same organ, while the same C/EBP can exert distinct functions in different organs. Modulating C/EBP expression or activity could regulate various molecular processes to alleviate fibrosis in multiple organs; therefore, novel C/EBPs-based therapeutic methods for treating fibrosis have attracted considerable attention. In this review, we will explore the features of C/EBPs and their critical functions in fibrosis in order to highlight new avenues for the development of novel therapies targeting C/EBPs.
Collapse
Affiliation(s)
- Lexun Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiaojiao Feng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanyue Deng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qianqian Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Quxing Wei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dewei Ye
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
6
|
Gligorijević N, Minić S, Nedić O. Structural changes of proteins in liver cirrhosis and consequential changes in their function. World J Gastroenterol 2022; 28:3780-3792. [PMID: 36157540 PMCID: PMC9367231 DOI: 10.3748/wjg.v28.i29.3780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/07/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
The liver is the site of synthesis of the majority of circulating proteins. Besides initial polypeptide synthesis, sophisticated machinery is involved in the further processing of proteins by removing parts of them and/or adding functional groups and small molecules tailoring the final molecule to suit its physiological purpose. Posttranslational modifications (PTMs) design a network of molecules with the common protein ancestor but with slightly or considerably varying activity/localization/purpose. PTMs can change under pathological conditions, giving rise to aberrant or overmodified proteins. Undesired changes in the structure of proteins most often accompany undesired changes in their function, such as reduced activity or the appearance of new effects. Proper protein processing is essential for the reactions in living beings and crucial for the overall quality control. Modifications that occur on proteins synthesized in the liver whose PTMs are cirrhosis-related are oxidation, nitration, glycosylation, acetylation, and ubiquitination. Some of them predominantly affect proteins that remain in liver cells, whereas others predominantly occur on proteins that leave the liver or originate from other tissues and perform their function in the circulation. Altered PTMs of certain proteins are potential candidates as biomarkers of liver-related diseases, including cirrhosis. This review will focus on PTMs on proteins whose structural changes in cirrhosis exert or are suspected to exert the most serious functional consequences.
Collapse
Affiliation(s)
- Nikola Gligorijević
- Department of Metabolism, University of Belgrade-Institute for the Application of Nuclear Energy, Belgrade 11080, Serbia
| | - Simeon Minić
- Centre of Excellence for Molecular Food Sciences and Department of Biochemistry, University of Belgrade-Faculty of Chemistry, Belgrade 11000, Serbia
| | - Olgica Nedić
- Department of Metabolism, University of Belgrade-Institute for the Application of Nuclear Energy, Belgrade 11080, Serbia
| |
Collapse
|
7
|
Caligiuri A, Gentilini A, Pastore M, Gitto S, Marra F. Cellular and Molecular Mechanisms Underlying Liver Fibrosis Regression. Cells 2021; 10:cells10102759. [PMID: 34685739 PMCID: PMC8534788 DOI: 10.3390/cells10102759] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic liver injury of different etiologies may result in hepatic fibrosis, a scar formation process consisting in altered deposition of extracellular matrix. Progression of fibrosis can lead to impaired liver architecture and function, resulting in cirrhosis and organ failure. Although fibrosis was previous thought to be an irreversible process, recent evidence convincingly demonstrated resolution of fibrosis in different organs when the cause of injury is removed. In the liver, due to its high regenerative ability, the extent of fibrosis regression and reversion to normal architecture is higher than in other tissues, even in advanced disease. The mechanisms of liver fibrosis resolution can be recapitulated in the following main points: removal of injurious factors causing chronic hepatic damage, elimination, or inactivation of myofibroblasts (through various cell fates, including apoptosis, senescence, and reprogramming), inactivation of inflammatory response and induction of anti-inflammatory/restorative pathways, and degradation of extracellular matrix. In this review, we will discuss the major cellular and molecular mechanisms underlying the regression of fibrosis/cirrhosis and the potential therapeutic approaches aimed at reversing the fibrogenic process.
Collapse
|
8
|
Lv X, Qiu J, Hao T, Zhang H, Jiang H, Tan Y. HDAC inhibitor Trichostatin A suppresses adipogenesis in 3T3-L1 preadipocytes. Aging (Albany NY) 2021; 13:17489-17498. [PMID: 34232916 PMCID: PMC8312440 DOI: 10.18632/aging.203238] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/04/2021] [Indexed: 12/30/2022]
Abstract
Background and purpose: Obesity is becoming a major global health issue and is mainly induced by the accumulation of adipose tissues mediated by adipogenesis, which is reported to be regulated by peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT enhancer-binding protein α (C/EBPα). Trichostatin A (TSA) is a novel histone deacetylase inhibitor (HDACI) that was recently reported to exert multiple pharmacological functions. The present study will investigate the inhibitory effect of TSA on adipogenesis, as well as the underlying mechanism. Methods: The adipogenesis of 3T3-L1 cells was induced by stimulation with a differentiation cocktail (DMI) medium for 8 days. MTT assay was used to measure the cell viability and Oil Red O staining was used to evaluate the adipogenesis of 3T3-L1 cells. The total level of triglyceride and released glycerol were detected to evaluate the lipolysis during 3T3-L1 adipogenesis. The expression levels of Leptin, fatty acid-binding protein 4 (FABP4), and sterol regulatory element-binding protein (SREBP1C) were determined by qRT-PCR. qRT-PCR assay was utilized to detect the expression levels of PPARγ and C/EBPα in 3T3-L1 cells. A high-fat diet (HFD) was used to construct an obese mice model, followed by the treatment with TSA. HE staining was conducted to evaluate the pathological state of adipose tissues. Body weights and the weights of adipose tissues were recorded to evaluate the anti-obesity property of TSA. Results: Firstly, the promoted lipid accumulation induced by DMI incubation was significantly reversed by the treatment with TSA in a dose-dependent manner. The elevated expression levels of Leptin, FABP4, SREBP1C, PPARγ, and C/EBPα induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Secondly, the inhibitory effect of TSA against the expression level of PPARγ and lipid accumulation was greatly abolished by an AMPK inhibitor. Lastly, the increased body weights and visceral adipocyte tissue weight, as well as the enlarged size of adipocytes induced by HFD were pronouncedly reversed by the administration of TSA. Conclusion: TSA inhibited adipogenesis in 3T3-L1 preadipocytes by activating the AMPK pathway.
Collapse
Affiliation(s)
- Xin Lv
- Department of Clinical Nutrition, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China.,Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Jun Qiu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Tao Hao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Haoran Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Haiping Jiang
- Department of Clinical Nutrition, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China.,Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Yang Tan
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518000, Guangdong, China
| |
Collapse
|
9
|
Hou C, Lu S, Su Y, Ding D, Tao L, Wang M, Wang Y, Liu X. C/EBP-α induces autophagy by binding to Beclin1 through its own acetylation modification in activated hepatic stellate cells. Exp Cell Res 2021; 405:112721. [PMID: 34217716 DOI: 10.1016/j.yexcr.2021.112721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/07/2021] [Accepted: 06/28/2021] [Indexed: 01/31/2023]
Abstract
The activation of hepatic stellate cells (HSCs) plays a key role in the occurrence of liver fibrosis,and promoting the apoptosis of activated HSCs or reducing the number of activated HSCs can reverse the development of liver fibrosis. In our previous studies, we have demonstrated that the CCAAT/enhancer binding protein α (C/EBP-α) played an important role in promoting the apoptosis of activated HSCs, thereby exerting an anti-liver fibrosis effect. Unlike apoptosis, autophagy, as a caspase-independent programmed cell death, can promptly remove the abnormal accumulation of substances or damaged organelles in cells and play a key role in regulating the homeostasis of intracellular environment. However, it is still unclear whether C/EBP-α participates in the occurrence of autophagy in HSCs. Therefore, in this study, we firstly used the methods of Western blot and immunofluorescence to characterize the consequence of C/EBP-α overexpression on the expression of proteins LC3B, P62, ATG5 and Beclin1 which were related to autophagy in HSCs. Subsequently, we performed Western blot and site-directed mutagenesis methods to clarify the type and related mechanism of autophagy which was induced by C/EBP-α. Here we show that C/EBP-α promotes the occurrence of autophagy in HSCs and the autophagy induced by C/EBP-α belongs to mitophagy. The stability of C/EBP-α protein regulates the level of autophagy in HSCs. In addition, acetylation of C/EBP-α also regulates the occurrence of autophagy in HSCs. Acetylation of lysine at positions K298, K302 and K326 of C/EBP-α promotes its binding to Beclin1. In conclusion, our study uncovers the role of C/EBP-α in regulating autophagy in HSCs, thereby providing a new strategy for clinical treatment of liver fibrosis.
Collapse
Affiliation(s)
- Chenjian Hou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, PR China
| | - Shan Lu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, PR China
| | - Ying Su
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, PR China
| | - Di Ding
- Department of Pathology, The Children's Hospital, Fudan University, Shanghai, 201102, China
| | - Lili Tao
- Department of Pathology, Peking University, Shenzhen Hospital, Shenzhen, 518036, China
| | - Meili Wang
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 201100, China
| | - Yuxiang Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, PR China.
| | - Xiuping Liu
- Department of Pathology, School of Basic Medical Sciences, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
10
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 36:299-309. [PMID: 34773731 DOI: 10.1515/dmpt-2020-0186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- College of Pharmacy, Arab Academy for Science, Technology & Maritime Transport, Alexandria, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
11
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 0:dmdi-2020-0186. [PMID: 33818027 DOI: 10.1515/dmdi-2020-0186] [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: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
12
|
Ganguly N, Chakrabarti S. Role of long non‑coding RNAs and related epigenetic mechanisms in liver fibrosis (Review). Int J Mol Med 2021; 47:23. [PMID: 33495817 PMCID: PMC7846421 DOI: 10.3892/ijmm.2021.4856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is one of the major liver pathologies affecting patients worldwide. It results from an improper tissue repair process following liver injury or inflammation. If left untreated, it ultimately leads to liver cirrhosis and liver failure. Long non‑coding RNAs (lncRNAs) have been implicated in a wide variety of diseases. They can regulate gene expression and modulate signaling. Some of the lncRNAs promote, while others inhibit liver fibrosis. Similarly, other epigenetic processes, such as methylation and acetylation regulate gene transcription and can modulate gene expression. Notably, there are several regulatory associations of lncRNAs with other epigenetic processes. A major mechanism of action of long non‑coding RNAs is to competitively bind to their target microRNAs (miRNAs or miRs), which in turn affects miRNA availability and bioactivity. In the present review, the role of lncRNAs and related epigenetic processes contributing to liver fibrosis is discussed. Finally, various potential therapeutic approaches targeting lncRNAs and related epigenetic processes, which are being considered as possible future treatment targets for liver fibrosis are identified.
Collapse
Affiliation(s)
- Niladri Ganguly
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
13
|
Claveria-Cabello A, Colyn L, Arechederra M, Urman JM, Berasain C, Avila MA, Fernandez-Barrena MG. Epigenetics in Liver Fibrosis: Could HDACs be a Therapeutic Target? Cells 2020; 9:cells9102321. [PMID: 33086678 PMCID: PMC7589994 DOI: 10.3390/cells9102321] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic liver diseases (CLD) represent a worldwide health problem. While CLDs may have diverse etiologies, a common pathogenic denominator is the presence of liver fibrosis. Cirrhosis, the end-stage of CLD, is characterized by extensive fibrosis and is markedly associated with the development of hepatocellular carcinoma. The most important event in hepatic fibrogenesis is the activation of hepatic stellate cells (HSC) following liver injury. Activated HSCs acquire a myofibroblast-like phenotype becoming proliferative, fibrogenic, and contractile cells. While transient activation of HSCs is part of the physiological mechanisms of tissue repair, protracted activation of a wound healing reaction leads to organ fibrosis. The phenotypic changes of activated HSCs involve epigenetic mechanisms mediated by non-coding RNAs (ncRNA) as well as by changes in DNA methylation and histone modifications. During CLD these epigenetic mechanisms become deregulated, with alterations in the expression and activity of epigenetic modulators. Here we provide an overview of the epigenetic alterations involved in fibrogenic HSCs transdifferentiation with particular focus on histones acetylation changes. We also discuss recent studies supporting the promising therapeutic potential of histone deacetylase inhibitors in liver fibrosis.
Collapse
Affiliation(s)
- Alex Claveria-Cabello
- Program of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (A.C.-C.); (L.C.); (M.A.); (C.B.)
| | - Leticia Colyn
- Program of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (A.C.-C.); (L.C.); (M.A.); (C.B.)
| | - Maria Arechederra
- Program of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (A.C.-C.); (L.C.); (M.A.); (C.B.)
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), 28029 Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain;
| | - Jesus M. Urman
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain;
- Department of Gastroenterology and Hepatology, Navarra University Hospital Complex, 31008 Pamplona, Spain
| | - Carmen Berasain
- Program of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (A.C.-C.); (L.C.); (M.A.); (C.B.)
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), 28029 Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain;
| | - Matias A. Avila
- Program of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (A.C.-C.); (L.C.); (M.A.); (C.B.)
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), 28029 Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain;
- Correspondence: (M.A.A.); (M.G.F.-B.); Tel.: +34-94-819-4700 (M.A.A.); +34-94-819-4700 (M.G.F.-B.)
| | - Maite G. Fernandez-Barrena
- Program of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (A.C.-C.); (L.C.); (M.A.); (C.B.)
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), 28029 Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain;
- Correspondence: (M.A.A.); (M.G.F.-B.); Tel.: +34-94-819-4700 (M.A.A.); +34-94-819-4700 (M.G.F.-B.)
| |
Collapse
|
14
|
Gao J, Yang J, Yu W, Hao R, Fan J, Wei J. Gooseberry anthocyanins protect mice hepatic fibrosis by inhibiting TGF-β/Smad pathway. FOOD BIOSCI 2020. [DOI: 10.1016/j.fbio.2020.100717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
15
|
Pan Q, Guo CJ, Xu QY, Wang JZ, Li H, Fang CH. miR-16 integrates signal pathways in myofibroblasts: determinant of cell fate necessary for fibrosis resolution. Cell Death Dis 2020; 11:639. [PMID: 32801294 PMCID: PMC7429878 DOI: 10.1038/s41419-020-02832-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/14/2022]
Abstract
Liver fibrosis is characterized by the transdifferentiation of hepatic stellate cells (HSCs) to myofibroblasts and poor response to treatment. This can be attributed to the myofibroblast-specific resistance to phenotype reversal. In this study, we complemented miR-16 into miR-16-deficient myofibroblasts and analyzed the global role of miR-16 using transcriptome profiling and generating a pathway-based action model underlying transcriptomic regulation. Phenotypic analysis of myofibroblasts and fibrogenic characterization were used to understand the effect of miR-16 on phenotypic remodeling of myofibroblasts. miR-16 expression altered the transcriptome of myofibroblasts to resemble that of HSCs. Simultaneous targeting of Smad2 and Wnt3a, etc. by miR-16 integrated signaling pathways of TGF-β and Wnt, etc., which underlay the comprehensive regulation of transcriptome. The synergistic effect of miR-16 on the signaling pathways abolished the phenotypic characteristics of myofibroblasts, including collagen production and inhibition of adipogenesis. In vivo, myofibroblast-specific expression of miR-16 not only eliminated mesenchymal cells with myofibroblast characteristics but also restored the phenotype of HSCs in perisinusoidal space. This phenotypic remodeling resolved liver fibrosis induced by chronic wound healing. Therefore, miR-16 may integrate signaling pathways crucial for the fate determination of myofibroblasts. Its global effect induces the reversal of HSC-to-myofibroblast transdifferentiation and, subsequently, the resolution of fibrogenesis. Taken together, these findings highlight the potential of miR-16 as a promising therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Qin Pan
- Department of Gastroenterology, Xin-Hua Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200092, China.
| | - Can-Jie Guo
- Department of Gastroenterology, Ren-Ji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200001, China
| | - Qing-Yang Xu
- Department of Gastroenterology, Xin-Hua Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200092, China
| | - Jin-Zhi Wang
- Department of Gastroenterology, Xin-Hua Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200092, China
| | - Han Li
- Department of Gastroenterology, Xin-Hua Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200092, China
| | - Chun-Hua Fang
- School of Electronics and Information Engineering, Tong-Ji University, Shanghai, 201804, China
| |
Collapse
|
16
|
Gao Q, Wei A, Chen F, Chen X, Ding W, Ding Z, Wu Z, Du R, Cao W. Enhancing PPARγ by HDAC inhibition reduces foam cell formation and atherosclerosis in ApoE deficient mice. Pharmacol Res 2020; 160:105059. [PMID: 32621955 DOI: 10.1016/j.phrs.2020.105059] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerosis (AS) is a risky cardiovascular disease with limited treatment options. Various pan or type-selective histone deacetylase (HDAC) inhibitors are reportedly atheroprotective against atherosclerosis (AS); however, the key effectors and the main cellular processes that mediate the protective effects remain poorly defined. Here, we report that PPARγ (Peroxisome proliferator-activated receptor gamma), a transcription factor actively involved in lipid metabolism with strong tissue protective and anti-inflammation properties, is a critical mediator of the anti-AS effects by HDAC inhibition. We showed that a well-known pan-HDAC inhibitor TSA (Trichostatin A) reduced foam cell formation of macrophages that is accompanied by a marked elevation of PPARγ and its downstream cholesterol efflux transporter ABCA1 (ATP-binding membrane cassette transport protein A1) and ABCG1. In an AS model of ApoE-/- mice fed on high-fat diet, TSA treatment alleviated AS lesions, similarly increased PPARγ and the downstream cholesterol transporters and mitigated the induction of inflammatory cytokine TNFα and IL-1β. Exploring the potential cause of PPARγ elevation revealed that TSA induced the acetylation of C/EBPα (CCAAT enhancer binding protein alpha), the upstream regulator of PPARγ, through which it increased PPARγ transactivation. More importantly, we generated a strain of PPARγ/ApoE double knockout mice and demonstrated that lack of PPARγ abrogated the protective effects of TSA on foam cell formation of peritoneal macrophages and the AS pathogenesis. Taken together, these results unravel that C/EBPα and PPARγ are the HDAC-sensitive components of an epigenetic signaling pathway mediating foam cell formation and AS development, and suggest that targeting C/EBPα/PPARγ axis by HDAC inhibitors possesses therapeutic potentials in retarding the progression of AS and the related cardiovascular diseases.
Collapse
Affiliation(s)
- Qi Gao
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Ai Wei
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Fang Chen
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Xingren Chen
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Wenwen Ding
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Zhiquan Ding
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Zhiwei Wu
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Ronghui Du
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China.
| | - Wangsen Cao
- Center for Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China.
| |
Collapse
|
17
|
Suh SH, Choi HS, Kim CS, Kim IJ, Cha H, Cho JM, Ma SK, Kim SW, Bae EH. CG200745, a Novel HDAC Inhibitor, Attenuates Kidney Fibrosis in a Murine Model of Alport Syndrome. Int J Mol Sci 2020; 21:ijms21041473. [PMID: 32098220 PMCID: PMC7073208 DOI: 10.3390/ijms21041473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/14/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Histone deacetylases have been a target of therapy for organ fibrosis. Here, we report the protective effect of CG200745 (CG), a novel histone deacetylase inhibitor, on tubulointerstitial fibrosis in Col4a3-/- mice, a murine model of Alport syndrome. Morphological analyses revealed CG treatment markedly alleviated kidney fibrosis in Col4a3-/- mice at the age of 7 weeks. CG prevented the activation of transforming growth factor β (TGFβ) and its downstream SMAD signaling in the kidney of Col4a3-/- mice. As critical upstream regulators of TGFβ signaling, immunoblotting of whole kidney lysate of Col4a3-/- mice reveled that intra-renal renin-angiotensin system (RAS) was activated with concurrent upregulation of inflammation and apoptosis, which were effectively suppressed by CG treatment. CG suppressed both activation of RAS and up-regulation of TGFβ signals in angiotensin II-stimulated HK-2 cells, a human kidney proximal tubular epithelial cell line. CG inhibited activation of TGFβ-driven signals and fibrosis in NRK-49F cells, a rat kidney fibroblast cell line, under angiotensin II-rich conditions. Collectively, CG was found to be effective both in proximal tubular epithelial cells by inhibiting local RAS and TGFβ signaling activation, as well as in fibroblasts by blocking their transition to myofibroblasts, attenuating renal fibrosis in a murine model of Alport syndrome.
Collapse
Affiliation(s)
- Sang Heon Suh
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Korea; (S.H.S.); (H.S.C.); (C.S.K.); (I.J.K.); (S.K.M.)
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju 61469, Korea
| | - Hong Sang Choi
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Korea; (S.H.S.); (H.S.C.); (C.S.K.); (I.J.K.); (S.K.M.)
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju 61469, Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Korea; (S.H.S.); (H.S.C.); (C.S.K.); (I.J.K.); (S.K.M.)
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju 61469, Korea
| | - In Jin Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Korea; (S.H.S.); (H.S.C.); (C.S.K.); (I.J.K.); (S.K.M.)
| | - Hyunju Cha
- Crystal Genomics, Inc., 5 F, Bldg A, Korea Bio Park, Seongnam 13488, Korea; (H.C.); (J.M.C.)
| | - Joong Myung Cho
- Crystal Genomics, Inc., 5 F, Bldg A, Korea Bio Park, Seongnam 13488, Korea; (H.C.); (J.M.C.)
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Korea; (S.H.S.); (H.S.C.); (C.S.K.); (I.J.K.); (S.K.M.)
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju 61469, Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Korea; (S.H.S.); (H.S.C.); (C.S.K.); (I.J.K.); (S.K.M.)
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju 61469, Korea
- Correspondence: (S.W.K.); (E.H.B.); Tel.: +82-62-220-6503 (S.W.K. & E.H.B.)
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju 61469, Korea; (S.H.S.); (H.S.C.); (C.S.K.); (I.J.K.); (S.K.M.)
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju 61469, Korea
- Correspondence: (S.W.K.); (E.H.B.); Tel.: +82-62-220-6503 (S.W.K. & E.H.B.)
| |
Collapse
|
18
|
Zhang Y, Distler JHW. Therapeutic molecular targets of SSc-ILD. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2020; 5:17-30. [DOI: 10.1177/2397198319899013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Systemic sclerosis is a fibrosing chronic connective tissue disease of unknown etiology. A major hallmark of systemic sclerosis is the uncontrolled and persistent activation of fibroblasts, which release excessive amounts of extracellular matrix, lead to organ dysfunction, and cause high mobility and motility of patients. Systemic sclerosis–associated interstitial lung disease is one of the most common fibrotic organ manifestations in systemic sclerosis and a major cause of death. Treatment options for systemic sclerosis–associated interstitial lung disease and other fibrotic manifestations, however, remain very limited. Thus, there is a huge medical need for effective therapies that target tissue fibrosis, vascular alterations, inflammation, and autoimmune disease in systemic sclerosis–associated interstitial lung disease. In this review, we discuss data suggesting therapeutic ways to target different genes in distinct tissues/organs that contribute to the development of SSc.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jörg HW Distler
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
19
|
Iacob S, Iacob DG. Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis. Front Microbiol 2019; 10:1676. [PMID: 31447793 PMCID: PMC6692454 DOI: 10.3389/fmicb.2019.01676] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
The ecosystem of the gut microbiota consists of diverse intestinal species with multiple metabolic and immunologic activities and it is closely connected with the intestinal epithelia and mucosal immune response, with which it builds a complex barrier against intestinal pathogenic bacteria. The microbiota ensures the integrity of the gut barrier through multiple mechanisms, either by releasing antibacterial molecules (bacteriocins) and anti-inflammatory short-chain fatty acids or by activating essential cell receptors for the immune response. Experimental studies have confirmed the role of the intestinal microbiota in the epigenetic modulation of the gut barrier through posttranslational histone modifications and regulatory mechanisms induced by epithelial miRNA in the epithelial lumen. Any quantitative or functional changes of the intestinal microbiota, referred to as dysbiosis, alter the immune response, decrease epithelial permeability and destabilize intestinal homeostasis. Consequently, the overgrowth of pathobionts (Staphylococcus, Pseudomonas, and Escherichia coli) favors intestinal translocations with Gram negative bacteria or their endotoxins and could trigger sepsis, septic shock, secondary peritonitis, or various intestinal infections. Intestinal infections also induce epithelial lesions and perpetuate the risk of bacterial translocation and dysbiosis through epithelial ischemia and pro-inflammatory cytokines. Furthermore, the decline of protective anaerobic bacteria (Bifidobacterium and Lactobacillus) and inadequate release of immune modulators (such as butyrate) affects the release of antimicrobial peptides, de-represses microbial virulence factors and alters the innate immune response. As a result, intestinal germs modulate liver pathology and represent a common etiology of infections in HIV immunosuppressed patients. Antibiotic and antiretroviral treatments also promote intestinal dysbiosis, followed by the selection of resistant germs which could later become a source of infections. The current article addresses the strong correlations between the intestinal barrier and the microbiota and discusses the role of dysbiosis in destabilizing the intestinal barrier and promoting infectious diseases.
Collapse
Affiliation(s)
- Simona Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Balş", Bucharest, Romania
| | - Diana Gabriela Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
20
|
Barcena-Varela M, Colyn L, Fernandez-Barrena MG. Epigenetic Mechanisms in Hepatic Stellate Cell Activation During Liver Fibrosis and Carcinogenesis. Int J Mol Sci 2019; 20:E2507. [PMID: 31117267 PMCID: PMC6566358 DOI: 10.3390/ijms20102507] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/17/2019] [Accepted: 05/19/2019] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is an essential component of chronic liver disease (CLD) and hepatocarcinogenesis. The fibrotic stroma is a consequence of sustained liver damage combined with exacerbated extracellular matrix (ECM) accumulation. In this context, activation of hepatic stellate cells (HSCs) plays a key role in both initiation and perpetuation of fibrogenesis. These cells suffer profound remodeling of gene expression in this process. This review is focused on the epigenetic alterations participating in the transdifferentiation of HSCs from the quiescent to activated state. Recent advances in the field of DNA methylation and post-translational modifications (PTM) of histones (acetylation and methylation) patterns are discussed here, together with altered expression and activity of epigenetic remodelers. We also consider recent advances in translational approaches, including the use of epigenetic marks as biomarkers and the promising antifibrotic properties of epigenetic drugs that are currently being used in patients.
Collapse
Affiliation(s)
| | - Leticia Colyn
- Hepatology Program, CIMA, University of Navarra, 31180 Pamplona, Spain.
| | - Maite G Fernandez-Barrena
- Hepatology Program, CIMA, University of Navarra, 31180 Pamplona, Spain.
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, 31180 Pamplona, Spain.
| |
Collapse
|
21
|
Zhao Y, Yu T, Zhang N, Chen J, Zhang P, Li S, Luo L, Cui Z, Qin Y, Liu F. Nuclear E-Cadherin Acetylation Promotes Colorectal Tumorigenesis via Enhancing β-Catenin Activity. Mol Cancer Res 2018; 17:655-665. [PMID: 30401720 DOI: 10.1158/1541-7786.mcr-18-0637] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/23/2018] [Accepted: 10/18/2018] [Indexed: 11/16/2022]
Abstract
The E-cadherin/β-catenin signaling pathway plays a critical role in the maintenance of epithelial architecture and regulation of tumor progression. Normally, E-cadherin locates on the cell surface with its cytosolic domain linking to the actin cytoskeleton through interaction with catenins. Although the nuclear localization of E-cadherin has been frequently observed in various types of cancers, little is known regarding the functional consequences of its nuclear translocation. Here, we showed that in colorectal cancer samples and cell lines, E-cadherin localized in the nucleus; and the nuclear localization was mediated through protein interaction with CTNND1. In the nucleus, E-cadherin was acetylated by CREB-binding protein at Lysine870 and Lysine871 in its β-catenin-binding domain, and the acetylation can be reversed by SIRT2. Acetylation of nuclear E-cadherin attenuated its interaction with β-catenin, which therefore released β-catenin from the complex, resulting in increased expression of its downstream genes and accelerated tumor growth and migration. Further study showed that acetylation level of nuclear E-cadherin had high prognostic significance in clinical colorectal samples. Taken together, our findings reveal a novel mechanism of tumor progression through posttranslational modification of E-cadherin, which may serve as a potential drug target of tumor therapy. IMPLICATIONS: This finding that acetylation of nuclear E-cadherin regulates β-catenin activity expands our understanding of the acetylation of E-cadherin promotes colorectal cancer cell growth and suggests novel therapeutic approaches of targeting acetylation in tumors.
Collapse
Affiliation(s)
- Yongxu Zhao
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Tao Yu
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Nan Zhang
- Department of Oncology, Jinan Central Hospital afflicted to Shandong University, Shandong, P.R. China
| | - Jianxia Chen
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, P.R. China
| | - Peng Zhang
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Shuang Li
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China
| | - Lijun Luo
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Department of Periodontology, School of Stomatology, Tongji University, Shanghai, P.R. China
| | - Zhenling Cui
- Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, P.R. China
| | - Yue Qin
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China.
| | - Feng Liu
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, P.R. China. .,Department of Oncology, Jinan Central Hospital afflicted to Shandong University, Shandong, P.R. China.,Shanghai Key Laboratory of Mycobacterium Tuberculosis, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, P.R. China
| |
Collapse
|
22
|
Inhibiting Arginine Methylation as a Tool to Investigate Cross-Talk with Methylation and Acetylation Post-Translational Modifications in a Glioblastoma Cell Line. Proteomes 2018; 6:proteomes6040044. [PMID: 30347783 PMCID: PMC6313862 DOI: 10.3390/proteomes6040044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/02/2018] [Accepted: 10/17/2018] [Indexed: 12/18/2022] Open
Abstract
Glioblastomas (GBM) are the most common grade 4 brain tumours; patients have very poor prognosis with an average survival of 15 months after diagnosis. Novel research lines have begun to explore aberrant protein arginine methylation (ArgMe) as a possible therapeutic target in GBM and ArgMe inhibitors are currently in clinical trials. Enzymes known as protein arginine methyltransferases (PRMT1-9) can lead to mono- or di-ArgMe, and in the latter case symmetric or asymmetric dimethylation (SDMA and ADMA, respectively). Using the most common GBM cell line, we have profiled the expression of PRMTs, used ArgMe inhibitors as tools to investigate post-translational modifications cross-talk and measured the effect of ArgMe inhibitors on cell viability. We have identified novel SDMA events upon inhibition of ADMA in GBM cells and spheroids. We have observed cross-talk between ADMA and lysine acetylation in GBM cells and platelets. Treatment of GBM cells with furamidine, a PRMT1 inhibitor, reduces cell viability in 2D and 3D models. These data provide new molecular understanding of a disease with unmet clinical needs.
Collapse
|