1
|
Liang C, Liu J, Jiang M, Zhu Y, Dong P. The advancement of targeted regulation of hepatic stellate cells using traditional Chinese medicine for the treatment of liver fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119298. [PMID: 39798676 DOI: 10.1016/j.jep.2024.119298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 01/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver fibrosis, which is a precursor to cirrhosis in chronic liver diseases, is driven by various factors. The activation and proliferation of hepatic stellate cells (HSCs) are recognized as a crucial phase in the progression of liver fibrosis. Compared with western drug therapy, Traditional Chinese medicine (TCM) and herbal medicine not only have the advantages of multi-target and multi-pathways in the treatment of liver fibrosis, but also have high safety without toxic side effects. AIM OF THE REVIEW This paper aims to compile and analyze the active ingredients in TCM and their corresponding signaling pathways that target and modulate the phenotype of hepatic stellate cells, offering a potential treatment for hepatic fibrosis. METHODS The Literature information was obtained from the scientific databases PubMed, Web of Science and CNKI from January 2010 to June 2020 with the aim of elucidating the intrinsic mechanisms and roles of TCM and natural medicine in the treatment of LF. The search terms included "liver fibrosis" or "hepatic fibrosis", "traditional Chinese medicine" or "Chinese herbal medicine", "medicinal plant", "natural plant", and "herb". RESULTS We described the antifibrosis activity of TCM and natural medicine in LF based on different signaling pathways. Plant medicine and herbal formulas regulated the related gene and protein expression via pathways such as TGF-β/Smad, PI3K/AKT/mTOR, MAPK and Wnt/β-catenin, which inhibit the proliferation, apoptosis, autophagy and activation of HSCs. CONCLUSION By reviewing both domestic and international literature on TCM interventions in liver fibrosis, this study presents a thorough evaluation of recent research progress and the challenges faced in the clinical application of TCM for this condition. The goal is to lay a solid foundation for further in-depth studies and to strengthen the theoretical framework in this field. The inhibitory effect of TCM and natural medicine on fibrosis was reflected in multiple levels and multiple pathways, providing reasonable evidence for new drug development. To make TCM and natural medicine widely and flexibly used in clinical practice, the efficacy, safety and mechanism of action need more in-depth experimental research. It also seeks to provide a theoretical foundation for future research on targeted therapies for liver fibrosis and related diseases.
Collapse
Affiliation(s)
- Chen Liang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Jingjing Liu
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Meixiu Jiang
- The Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Yan Zhu
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Pengzhi Dong
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
2
|
Wen S, Chang S, Zhang H, Zhang W, Guo Y, Zhang N, Yang A, Sun Y, Liu Z. Shikonin modulates activated fibroblast apoptosis in silicosis fibrosis via the PI3K/Akt signaling pathway: A network pharmacology approach. Toxicol Appl Pharmacol 2025; 495:117236. [PMID: 39855310 DOI: 10.1016/j.taap.2025.117236] [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: 11/18/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Silicosis is a lung disease caused by the inhalation of free crystalline silica and is characterized mainly by lung inflammation and progressive pulmonary fibrosis. Shikonin, a biologically active compound isolated from the traditional Chinese medicine Comfrey, has been shown to have significant antifibrotic effects. However, the molecular mechanisms underlying the antifibrotic effects of SHK in silicosis remain unclear. METHODS This study used a combination of network pharmacology, molecular docking, molecular dynamics simulation, and in vitro experimental validation to investigate the potential targets of SHK in silicosis. RESULTS Network pharmacology analysis identified 208 cross genes associated with disease drugs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis show that these intersecting genes are significantly associated with the PI3K/Akt signaling pathway. Protein protein interaction (PPI) network analysis further revealed 10 core crossover genes, namely ALB, Akt1, STAT3, CASP3, EGFR, MMP9, Bcl-2, ESR1, HSP90AA1, and NF-κB1. Among them, Akt1 and Bcl-2 have the strongest binding ability to SHK. The in vitro experimental results showed that SHK can significantly inhibit the activation of fibroblasts and promote apoptosis of activated fibroblasts through the PI3K/Akt signaling pathway. CONCLUSION SHK alleviates silica induced silicosis fibrosis by inhibiting the transformation of fibroblasts into myofibroblasts through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Shengpeng Wen
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Sirong Chang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Huning Zhang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Wenyue Zhang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Yi Guo
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Na Zhang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China
| | - Anning Yang
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China; Key Laboratory of Metabolic Cardiovascular Disease Research, National Health Commission, Ningxia Medical University, Yinchuan, Ningxia 750000, China.
| | - Yue Sun
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China.
| | - Zhihong Liu
- School of Public Health, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750000, China.
| |
Collapse
|
3
|
Wu QJ, Chen LY, Sun QM, Wang N, Han D, Lv WL. Quantitative pharmacodynamics functional evaluation of Chinese medicine Qizhu formula in mice with dynamic near-infrared photoacoustic imaging. PHOTOACOUSTICS 2025; 41:100667. [PMID: 39640433 PMCID: PMC11615922 DOI: 10.1016/j.pacs.2024.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 12/07/2024]
Abstract
Background & Aims Effective anti-fibrotic drugs and new non-invasive evaluation methods for liver fibrosis (LF) are urgently needed. Our study aimed to evaluate the histological effects of the Qizhu (QZ) formula on LF and to explore a non-invasive Near-infrared photoacoustic imaging (NIR-PAI) kinetic model for liver function detection and pharmacodynamic evaluation. Methods C57BL/6 J mice were randomly divided into six groups (n=6). An LF model was induced by CCl4 for 8 weeks, followed by an 8-week treatment period. Histological and serological parameters were assessed, and indocyanine green (ICG) metabolism (maximum peak time [Tmax] and half-life [T1/2]) was monitored by NIR-PAI. Spearman correlation analysis was conducted to evaluate correlations. Results & Conclusions Histological and serological results confirmed the anti-fibrotic effects of QZ. NIR-PAI kinetic parameters indicated that QZ shortened the Tmax and T1/2 of ICG. There were good correlations between ICG metabolism and liver histopathology. The non-invasive NIR-PAI kinetic model shows potential in liver function detection and pharmacodynamic evaluation.
Collapse
Affiliation(s)
- Qing-Juan Wu
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Lan-Yu Chen
- Natl Ctr Nanosci & Technol, CAS Ctr Excellence Nanosci, Beijing, PR China
| | - Quan-Mei Sun
- Natl Ctr Nanosci & Technol, CAS Ctr Excellence Nanosci, Beijing, PR China
| | - Ning Wang
- Natl Ctr Nanosci & Technol, CAS Ctr Excellence Nanosci, Beijing, PR China
| | - Dong Han
- Natl Ctr Nanosci & Technol, CAS Ctr Excellence Nanosci, Beijing, PR China
| | - Wen-Liang Lv
- Guang 'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| |
Collapse
|
4
|
Duan H, Song S, Li R, Hu S, Zhuang S, Liu S, Li X, Gao W. Strategy for treating MAFLD: Electroacupuncture alleviates hepatic steatosis and fibrosis by enhancing AMPK mediated glycolipid metabolism and autophagy in T2DM rats. Diabetol Metab Syndr 2024; 16:218. [PMID: 39261952 PMCID: PMC11389443 DOI: 10.1186/s13098-024-01432-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/24/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Recent studies have highlighted type 2 diabetes (T2DM) as a significant risk factor for the development of metabolic dysfunction-associated fatty liver disease (MAFLD). This investigation aimed to assess electroacupuncture's (EA) impact on liver morphology and function in T2DM rats, furnishing experimental substantiation for its potential to stall MAFLD progression in T2DM. METHODS T2DM rats were induced by a high-fat diet and a single intraperitoneal injection of streptozotocin, and then randomly assigned to five groups: the T2DM group, the electroacupuncture group, the metformin group, combination group of electroacupuncture and metformin, combination group of electroacupuncture and Compound C. The control group received a standard diet alongside intraperitoneal citric acid - sodium citrate solution injections. After a 6-week intervention, the effects of each group on fasting blood glucose, lipids, liver function, morphology, lipid droplet infiltration, and fibrosis were evaluated. Techniques including Western blotting, qPCR, immunohistochemistry, and immunofluorescence were employed to gauge the expression of key molecules in AMPK-associated glycolipid metabolism, insulin signaling, autophagy, and fibrosis pathways. Additionally, transmission electron microscopy facilitated the observation of liver autophagy, lipid droplets, and fibrosis. RESULTS Our studies indicated that hyperglycemia, hyperlipidemia and IR promoted lipid accumulation, pathological and functional damage, and resulting in hepatic steatosis and fibrosis. Meanwhile, EA enhanced the activation of AMPK, which in turn improved glycolipid metabolism and autophagy through promoting the expression of PPARα/CPT1A and AMPK/mTOR pathway, inhibiting the expression of SREBP1c, PGC-1α/PCK2 and TGFβ1/Smad2/3 signaling pathway, ultimately exerting its effect on ameliorating hepatic steatosis and fibrosis in T2DM rats. The above effects of EA were consistent with metformin. The combination of EA and metformin had significant advantages in increasing hepatic AMPK expression, improving liver morphology, lipid droplet infiltration, fibrosis, and reducing serum ALT levels. In addition, the ameliorating effects of EA on the progression of MAFLD in T2DM rats were partly disrupted by Compound C, an inhibitor of AMPK. CONCLUSIONS EA upregulated hepatic AMPK expression, curtailing gluconeogenesis and lipogenesis while boosting fatty acid oxidation and autophagy levels. Consequently, it mitigated blood glucose, lipids, and insulin resistance in T2DM rats, thus impeding liver steatosis and fibrosis progression and retarding MAFLD advancement.
Collapse
Affiliation(s)
- Haoru Duan
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
- Department of Acupuncture and Moxibustion, Chaoyang District Traditional Chinese Medicine Hospital, Beijing, 100026, China
| | - Shanshan Song
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
- Department of Acupuncture and Moxibustion, China- Japan Friendship Hospital, Beijing, 100029, China
| | - Rui Li
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Suqin Hu
- Department of Gastroenterology, Henan Province Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Henan, 450002, China
| | - Shuting Zhuang
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shaoyang Liu
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaolu Li
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Gao
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
5
|
Li Z, Yang Y, Gao F. Monomeric compounds from natural products for the treatment of pulmonary fibrosis: a review. Inflammopharmacology 2024; 32:2203-2217. [PMID: 38724690 DOI: 10.1007/s10787-024-01485-0] [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: 01/09/2024] [Accepted: 04/23/2024] [Indexed: 08/06/2024]
Abstract
Pulmonary fibrosis (PF) is the end stage of lung injury and chronic lung diseases that results in diminished lung function, respiratory failure, and ultimately mortality. Despite extensive research, the pathogenesis of this disease remains elusive, and effective therapeutic options are currently limited, posing a significant clinical challenge. In addition, research on traditional Chinese medicine and naturopathic medicine is hampered by several complications due to complex composition and lack of reference compounds. Natural product monomers, possessing diverse biological activities and excellent safety profiles, have emerged as potential candidates for preventing and treating PF. The effective anti-PF ingredients identified can be generally divided into flavonoids, saponins, polysaccharides, and alkaloids. Specifically, these monomeric compounds can attenuate inflammatory response, oxidative stress, and other physiopathological processes of the lung through many signaling pathways. They also improve pulmonary factors. Additionally, they ameliorate epithelial-mesenchymal transition (EMT) and fibroblast-myofibroblast transdifferentiation (FMT) by regulating multiple signal amplifiers in the lungs, thereby mitigating PF. This review highlights the significant role of monomer compounds derived from natural products in reducing inflammation, oxidative stress, and inhibiting EMT process. The article provides comprehensive information and serves as a solid foundation for further exploration of new strategies to harness the potential of botanicals in the treatment of PF.
Collapse
Affiliation(s)
- Zhuqing Li
- University of Shanghai for Science and Technology, 516, Jungong Road, Shanghai, 200093, China
| | - Yanyong Yang
- Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, China.
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, People's Republic of China.
| | - Fu Gao
- University of Shanghai for Science and Technology, 516, Jungong Road, Shanghai, 200093, China.
- Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, China.
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
6
|
Song Y, Ding Q, Hao Y, Cui B, Ding C, Gao F. Pharmacological Effects of Shikonin and Its Potential in Skin Repair: A Review. Molecules 2023; 28:7950. [PMID: 38138440 PMCID: PMC10745356 DOI: 10.3390/molecules28247950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Currently, skin injuries have a serious impact on people's lives and socio-economic stress. Shikonin, a naphthoquinone compound derived from the root of the traditional Chinese medicine Shikonin, has favorable biological activities such as anti-inflammatory, antibacterial, immunomodulatory, anticancer, and wound-healing-promoting pharmacological activities. It has been reported that Shikonin can be used for repairing skin diseases due to its wide range of pharmacological effects. Moreover, the antimicrobial activity of Shikonin can play a great role in food and can also reduce the number of pathogenic bacteria in food. This paper summarizes the research on the pharmacological effects of Shikonin in recent years, as well as research on the mechanism of action of Shikonin in the treatment of certain skin diseases, to provide certain theoretical references for the clinical application of Shikonin, and also to provides research ideas for the investigation of the mechanism of action of Shikonin in other skin diseases.
Collapse
Affiliation(s)
- Yanping Song
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology University, Jilin 132101, China;
| | - Qiteng Ding
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China;
| | - Yuewen Hao
- Jilin Jianwei Natural Biotechnology Co., Ltd., Linjiang 134600, China; (Y.H.); (B.C.)
| | - Bing Cui
- Jilin Jianwei Natural Biotechnology Co., Ltd., Linjiang 134600, China; (Y.H.); (B.C.)
| | - Chuanbo Ding
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology University, Jilin 132101, China;
- Jilin Aodong Yanbian Pharmaceutical Co., Ltd., Dunhua 133700, China
| | - Feng Gao
- College of Traditional Chinese Medicine, Jilin Agriculture Science and Technology University, Jilin 132101, China;
| |
Collapse
|
7
|
Liao X, Ruan X, Yao P, Yang D, Wu X, Zhou X, Jing J, Wei D, Liang Y, Zhang T, Qin S, Jiang H. LncRNA-Gm9866 promotes liver fibrosis by activating TGFβ/Smad signaling via targeting Fam98b. J Transl Med 2023; 21:778. [PMID: 37919785 PMCID: PMC10621198 DOI: 10.1186/s12967-023-04642-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE The exact mechanism and target molecules of liver fibrosis have remained largely elusive. Here, we investigated the role of long noncoding RNA Gm9866(lncRNA-Gm9866) on liver fibrosis. METHODS The transcription of lncRNA-Gm9866 in activated cells and mouse fibrotic livers was determined by quantitative polymerase chain reaction (qRT-PCR). The effects of lentivirus-mediated knockdown or overexpression of lncRNA-Gm9866 in liver fibrosis were examined in vitro and in vivo. Furthermore, bioinformatics analysis, cell samples validation, fluorescence in situ hybridization (FISH) co-localization, RNA binding protein immunoprecipitation (RIP), actinomycin D test and Western blot (WB) were carried out to explore the potential mechanism of lncRNA-Gm9866. RESULTS The expression of α-smooth muscle actin (α-SMA), Collagen I (COL-1) and lncRNA-Gm9866 were significantly increased in tissues and cells. Overexpressing lncRNA-Gm9866 promoted the activation of hepatic stellate cells (HSCs). Silencing lncRNA-Gm9866 inhibited the activation of HSCs and transforming growth factor-β1 (TGFβ1) induced fibrosis. Overexpressing lncRNA-Gm9866 promoted hepatocytes (HCs) apoptosis and the expression of pro-fibrogenic genes, inhibited the proliferation and migration of HCs. Knockdown of lncRNA-Gm9866 inhibited the apoptosis of HCs, the expression of pro-fibrogenic genes, TGFβ1 induced fibrosis and the occurrence of carbon tetrachloride (CCl4)-induced liver fibrosis, and promoted the proliferation and migration of HCs. Mechanistically, lncRNA-Gm9866 may directly bine with Fam98b. Silencing Fam98b in stably overexpressing lncRNA-Gm9866 cell lines reversed the increase of pro-fibrogenic genes and pro-apoptotic genes, fibrosis related pathway protein TGFβ1, Smad2/3, p-Smad2/3 and Notch3 induced by overexpressing lncRNA-Gm9866. CONCLUSIONS LncRNA-Gm9866 may regulate TGFβ/Smad and Notch pathways by targeting Fam98b to regulate liver fibrosis. LncRNA-Gm9866 may be a new target for diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Xiaomin Liao
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Xianxian Ruan
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Peishan Yao
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Dan Yang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xianbin Wu
- Department of Gastroenterology, The Wuming Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Xia Zhou
- Department of Emergency, People's Hospital of Guizhou Province, Guiyang, 550000, Guizhou, China
| | - Jie Jing
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Dafu Wei
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yaodan Liang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Taicheng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Shanyu Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China.
| | - Haixing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
8
|
Peng X, Yang H, Tao L, Xiao J, Zeng Y, Shen Y, Yu X, Zhu F, Qin J. Fluorofenidone alleviates liver fibrosis by inhibiting hepatic stellate cell autophagy via the TGF-β1/Smad pathway: implications for liver cancer. PeerJ 2023; 11:e16060. [PMID: 37790613 PMCID: PMC10542821 DOI: 10.7717/peerj.16060] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/17/2023] [Indexed: 10/05/2023] Open
Abstract
Objectives Liver fibrosis is a key stage in the progression of various chronic liver diseases to cirrhosis and liver cancer, but at present, there is no effective treatment. This study investigated the therapeutic effect of the new antifibrotic drug fluorofenidone (AKF-PD) on liver fibrosis and its related mechanism, providing implications for liver cancer. Materials and Methods The effects of AKF-PD on hepatic stellate cell (HSC) autophagy and extracellular matrix (ECM) expression were assessed in a carbon tetrachloride (CCl4)-induced rat liver fibrosis model. In vitro, HSC-T6 cells were transfected with Smad2 and Smad3 overexpression plasmids and treated with AKF-PD. The viability and number of autophagosomes in HSC-T6 cells were examined. The protein expression levels of Beclin-1, LC3 and P62 were examined by Western blotting. The Cancer Genome Atlas (TCGA) database was used for comprehensively analyzing the prognostic values of SMAD2 and SMAD3 in liver cancer. The correlation between SMAD2, SMAD3, and autophagy-related scores in liver cancer was explored. The drug prediction of autophagy-related scores in liver cancer was explored. Results AKF-PD attenuated liver injury and ECM deposition in the CCl4-induced liver fibrosis model. In vitro, the viability and number of autophagosomes in HSCs were reduced significantly by AKF-PD treatment. Meanwhile, the protein expression of FN, α-SMA, collagen III, Beclin-1 and LC3 was increased, and P62 was reduced by the overexpression of Smad2 and Smad3; however, AKF-PD reversed these effects. SMAD2 and SMAD3 were hazardous factors in liver cancer. SMAD2 and SMAD3 correlated with autophagy-related scores in liver cancer. Autophagy-related scores could predict drug response in liver cancer. Conclusions AKF-PD alleviates liver fibrosis by inhibiting HSC autophagy via the transforming growth factor (TGF)-β1/Smadpathway. Our study provided some implications about how liver fibrosis was connected with liver cancer by SMAD2/SMAD3 and autophagy.
Collapse
Affiliation(s)
- Xiongqun Peng
- Department of Gastroenterology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Huixiang Yang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephropathy, Xiangya Hospital, Central South University, Changsha, China
| | - Jingni Xiao
- Department of Nephrology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Ya Zeng
- Department of Gastroenterology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Yueming Shen
- Department of Gastroenterology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Xueke Yu
- Department of Gastroenterology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Fei Zhu
- Department of General Surgery, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Jiao Qin
- Department of Nephrology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
9
|
Siapoush S, Rezaei R, Alavifard H, Hatami B, Zali MR, Vosough M, Lorzadeh S, Łos MJ, Baghaei K, Ghavami S. Therapeutic implications of targeting autophagy and TGF-β crosstalk for the treatment of liver fibrosis. Life Sci 2023; 329:121894. [PMID: 37380126 DOI: 10.1016/j.lfs.2023.121894] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/19/2023] [Accepted: 06/25/2023] [Indexed: 06/30/2023]
Abstract
Liver fibrosis is characterized by the excessive deposition and accumulation of extracellular matrix components, mainly collagens, and occurs in response to a broad spectrum of triggers with different etiologies. Under stress conditions, autophagy serves as a highly conserved homeostatic system for cell survival and is importantly involved in various biological processes. Transforming growth factor-β1 (TGF-β1) has emerged as a central cytokine in hepatic stellate cell (HSC) activation and is the main mediator of liver fibrosis. A growing body of evidence from preclinical and clinical studies suggests that TGF-β1 regulates autophagy, a process that affects various essential (patho)physiological aspects related to liver fibrosis. This review comprehensively highlights recent advances in our understanding of cellular and molecular mechanisms of autophagy, its regulation by TGF-β, and the implication of autophagy in the pathogenesis of progressive liver disorders. Moreover, we evaluated crosstalk between autophagy and TGF-β1 signalling and discussed whether simultaneous inhibition of these pathways could represent a novel approach to improve the efficacy of anti-fibrotic therapy in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Samaneh Siapoush
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramazan Rezaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Helia Alavifard
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland; Autophagy Research Center, Department of Biochemistry; Shiraz University of Medical Sciences, Shiraz, Iran; LinkoCare Life Sciences AB, Linkoping, Sweden
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
10
|
Qu H, Liu J, Zhang D, Xie R, Wang L, Hong J. Glycolysis in Chronic Liver Diseases: Mechanistic Insights and Therapeutic Opportunities. Cells 2023; 12:1930. [PMID: 37566009 PMCID: PMC10417805 DOI: 10.3390/cells12151930] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Chronic liver diseases (CLDs) cover a spectrum of liver diseases, ranging from nonalcoholic fatty liver disease to liver cancer, representing a growing epidemic worldwide with high unmet medical needs. Glycolysis is a conservative and rigorous process that converts glucose into pyruvate and sustains cells with the energy and intermediate products required for diverse biological activities. However, abnormalities in glycolytic flux during CLD development accelerate the disease progression. Aerobic glycolysis is a hallmark of liver cancer and is responsible for a broad range of oncogenic functions including proliferation, invasion, metastasis, angiogenesis, immune escape, and drug resistance. Recently, the non-neoplastic role of aerobic glycolysis in immune activation and inflammatory disorders, especially CLD, has attracted increasing attention. Several key mediators of aerobic glycolysis, including HIF-1α and pyruvate kinase M2 (PKM2), are upregulated during steatohepatitis and liver fibrosis. The pharmacological inhibition or ablation of PKM2 effectively attenuates hepatic inflammation and CLD progression. In this review, we particularly focused on the glycolytic and non-glycolytic roles of PKM2 in the progression of CLD, highlighting the translational potential of a glycolysis-centric therapeutic approach in combating CLD.
Collapse
Affiliation(s)
| | | | | | | | | | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China; (H.Q.)
| |
Collapse
|
11
|
Liang D, Liu L, Zhao Y, Luo Z, He Y, Li Y, Tang S, Tang J, Chen N. Targeting extracellular matrix through phytochemicals: a promising approach of multi-step actions on the treatment and prevention of cancer. Front Pharmacol 2023; 14:1186712. [PMID: 37560476 PMCID: PMC10407561 DOI: 10.3389/fphar.2023.1186712] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023] Open
Abstract
Extracellular matrix (ECM) plays a pivotal and dynamic role in the construction of tumor microenvironment (TME), becoming the focus in cancer research and treatment. Multiple cell signaling in ECM remodeling contribute to uncontrolled proliferation, metastasis, immune evasion and drug resistance of cancer. Targeting trilogy of ECM remodeling could be a new strategy during the early-, middle-, advanced-stages of cancer and overcoming drug resistance. Currently nearly 60% of the alternative anticancer drugs are derived from natural products or active ingredients or structural analogs isolated from plants. According to the characteristics of ECM, this manuscript proposes three phases of whole-process management of cancer, including prevention of cancer development in the early stage of cancer (Phase I); prevent the metastasis of tumor in the middle stage of cancer (Phase II); provide a novel method in the use of immunotherapy for advanced cancer (Phase III), and present novel insights on the contribution of natural products use as innovative strategies to exert anticancer effects by targeting components in ECM. Herein, we focus on trilogy of ECM remodeling and the interaction among ECM, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), and sort out the intervention effects of natural products on the ECM and related targets in the tumor progression, provide a reference for the development of new drugs against tumor metastasis and recurrence.
Collapse
Affiliation(s)
- Dan Liang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunjie Zhao
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Zhenyi Luo
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Yadi He
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nianzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Liao X, Ruan X, Wu X, Deng Z, Qin S, Jiang H. Identification of Timm13 protein translocase of the mitochondrial inner membrane as a potential mediator of liver fibrosis based on bioinformatics and experimental verification. J Transl Med 2023; 21:188. [PMID: 36899394 PMCID: PMC9999505 DOI: 10.1186/s12967-023-04037-2] [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: 11/09/2022] [Accepted: 03/05/2023] [Indexed: 03/12/2023] Open
Abstract
OBJECTIVE To explore the association between translocase of the inner mitochondrial membrane 13 (Timm13) and liver fibrosis. METHODS Gene expression profiles of GSE167033 were collected from Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) between liver disease and normal samples were analyzed using GEO2R. Gene Ontology and Enrichment function were performed, a protein-protein interaction (PPI) network was constructed via the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), and the hub genes of the PPI network were calculated by MCODE plug-in in Cytoscape. We validated the transcriptional and post-transcriptional expression levels of the top correlated genes using fibrotic animal and cell models. A cell transfection experiment was conducted to silence Timm13 and detect the expression of fibrosis genes and apoptosis genes. RESULTS 21,722 genes were analyzed and 178 DEGs were identified by GEO2R analysis. The top 200 DEGs were selected and analyzed in STRING for PPI network analysis. Timm13 was one of the hub genes via the PPI network. We found that the mRNA levels of Timm13 in fibrotic liver tissue decreased (P < 0.05), and the mRNA and protein levels of Timm13 also decreased when hepatocytes were stimulated with transforming growth factor-β1. Silencing Timm13 significantly reduced the expression of profibrogenic genes and apoptosis related genes. CONCLUSIONS The results showed that Timm13 is closely related to liver fibrosis and silencing Timm13 significantly reduced the expression of profibrogenic genes and apoptosis related genes, which will provide novel ideas and targets for the clinical diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Xiaomin Liao
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Xianxian Ruan
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Xianbin Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Zhejun Deng
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Shanyu Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| | - Haixing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
13
|
Fu X, Chang J, Jiao D, Zhu M, Ma Y. SLIT3 knockdown inhibited TGF-β-induced hepatic stellate cells activation by down-regulating YAP signal. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-023-00336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
Objective
Liver fibrosis is a chronic liver disease caused by a variety of pathophysiological. However, there are no effective treatments to combat it. HSCs are a major source of fibrotic cells and exploring the mechanisms of HSC activation may provide new strategies for the treatment of liver fibrosis.
Objectives
To explore the role and underlying mechanism of SLIT3 in HSCs fibrosis.
Results
GSE163211 dataset analysis identified aberrant expression of SLIT3 in NASH F1-F4 tissues and SLIT3 expression level was positively correlated with fibrosis-related proteins. In vitro experiments showed that TGF-β induced upregulation of SLIT3 in LX-2 cells. Knockdown of SLIT3 significantly inhibited TGF-β-induced α-SMA, COL1A2, and COL1A1 expression, inhibited excessive cell proliferation and migration, and suppressed YAP activity.
Conclusion
Collectively, our findings suggest that SLIT3 deficiency alleviates TGF-β-induced HSCs activation by inhibiting YAP activity.
Collapse
|
14
|
Yu J, Li P, Duan Z, Liu X. Effect of Qiling Jiaogulan Powder on Pulmonary Fibrosis and Pulmonary Arteriole Remodeling in Low-Temperature-Exposed Broilers. Animals (Basel) 2022; 13:ani13010005. [PMID: 36611616 PMCID: PMC9817788 DOI: 10.3390/ani13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Chinese herbal medicine plays an important role in regulating the nutritional metabolism of poultry and maintaining or improving normal physiological functions and animal health. The present study investigated the effects of dietary supplementation with Qiling Jiaogulan Powder (QLJP) on pulmonary fibrosis and pulmonary arteriole remodeling in low temperature-exposed broilers. Seven-day-old Ross 308 broilers (n = 240) were reared adaptively to 14 days of age. The broilers were randomly divided into six groups: A control group (basal diet and normal feeding temperature); model group (basal diet); low-, medium- and high-dose QLJP groups (basal diet supplemented with 1 g/kg, 2 g/kg, 4 g/kg QLJP); and L-Arg group (basal diet supplemented with 10 g/kg L-arginine). Additionally, all the broilers, except the broilers in the control group, from the age of 14 days old, had a house temperature continuously lowered by 2 °C each day until it reached 12 °C at 21 days of age, and the low temperature was maintained until the end of the experiment. There were four replicates per group and 10 birds per replicate. The results showed that the structure of the lung tissue was clearer and basically intact in the broilers in the QLJP groups, with a small number of collagen fibers formed, and the content of hydroxyproline (HYP) was significantly reduced. QLJP improved pulmonary arteriole lesions, such as tunica media thickening, intimal hyperplasia, arterial wall hypertrophy, and lumen narrowing. QLJP reduced the relative media thickness (%) and relative medial area (%) of the pulmonary arteriole, and significantly decreased the expression level of the alpha-smooth muscle actin (α-SMA) protein in pulmonary arteriole, which alleviated pulmonary arteriole remodeling. The quantitative real-time PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA) results showed that QLJP treatment significantly reduced the gene and protein levels of transforming growth factor-beta l (TGF-β1) and Smad2 in the lung and downregulated the gene and protein levels of collagen type I alpha 1 (COL1A1) and matrix metalloproteinase 2 (MMP2). In conclusion, the results of our study suggested that dietary supplementation with QLJP improved pulmonary fibrosis and pulmonary arteriole remodeling by inhibiting the expression of genes related to the TGF-β1/Smad2 signaling pathway and inhibited the occurrence and development of pulmonary arterial hypertension in low-temperature-exposed broilers.
Collapse
Affiliation(s)
- Juan Yu
- School of Life Sciences and Basic Medicine, Xinxiang University, Xinxiang 453003, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030800, China
| | - Peng Li
- School of Life Sciences and Basic Medicine, Xinxiang University, Xinxiang 453003, China
| | - Zhibian Duan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030800, China
| | - Xingyou Liu
- School of Life Sciences and Basic Medicine, Xinxiang University, Xinxiang 453003, China
- Correspondence:
| |
Collapse
|
15
|
Rahman MM, Islam MR, Akash S, Shohag S, Ahmed L, Supti FA, Rauf A, Aljohani ASM, Al Abdulmonem W, Khalil AA, Sharma R, Thiruvengadam M. Naphthoquinones and derivatives as potential anticancer agents: An updated review. Chem Biol Interact 2022; 368:110198. [PMID: 36179774 DOI: 10.1016/j.cbi.2022.110198] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/03/2022]
Abstract
One of the leading global causes of death is cancer; even though several treatment methods have improved survival rates, the incidence and fatality rates remain high. Naphthoquinones are a type of quinone that is found in nature and has vital biological roles. These chemicals have anticancer (antineoplastic), analgesic, anti-inflammatory, antimalarial, antifungal, antiviral, antitrypanosomal, antischistosomal, leishmanicidal, and anti-ulcerative effects. Direct addition of a substituent group to the 1,4-naphthoquinone ring can alter the naphthoquinone's oxidation/reduction and acid/base characteristics, and the activity can be altered. Because of their pharmacological properties, such as anticancer activity and probable therapeutic application, naphthoquinones have greatly interested the scientific community. Some chemicals having a quinone ring in malignant cells have been found to have antiproliferative effects. Naphthoquinones' deadly impact is connected with the inhibition of electron transporters, the uncoupling of oxidative phosphorylation, the creation of ROS, and the formation of protein adducts, notably with -SH enzyme groups. This review article aims to discuss naphthoquinones and their derivatives, which act against cancer and their future perspectives. This review covers several studies highlighting the potent anticancer properties of naphthoquinones. Further, various proposed mechanisms of anticancer actions of naphthoquinones have been summarized in this review.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Sheikh Shohag
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Mirpur 12, Dhaka, 1216, Bangladesh
| | - Limon Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Fatema Akter Supti
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, 1207, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine Qassim University, Buraydah, Saudi Arabia
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Pakistan, Pakistan
| | - Rohit Sharma
- Department of Rasa Shastra & Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Sciences, Konkuk University, Seoul 05029, South Korea; Department of Microbiology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600077, India.
| |
Collapse
|
16
|
Zhang Y, Hua L, Lin C, Yuan M, Xu W, Raj D. A, Venkidasamy B, Cespedes-Acuna CL, Nile SH, Yan G, Zheng H. Pien-Tze-Huang alleviates CCl4-induced liver fibrosis through the inhibition of HSC autophagy and the TGF-β1/Smad2 pathway. Front Pharmacol 2022; 13:937484. [PMID: 36188553 PMCID: PMC9523731 DOI: 10.3389/fphar.2022.937484] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/26/2022] [Indexed: 11/28/2022] Open
Abstract
Ethnopharmacological relevance: Pien-Tze-Huang (PZH)—a traditional Chinese medicine (TCM) compound—has been employed to treat various liver inflammation and tumors for over 10 decades. Interestingly, most of the pharmacological effects had been validated and explored toward liver ailment along with pro-inflammatory conditions and cancer at the cellular and molecular level to date. Aim of the study: The present study aimed to investigate the therapeutic effect of PZH on autophagy and TGF-β1 signaling pathways in rats with liver fibrosis and hepatic stellate cell line (HSC). Materials and methods: Male SD rats with carbon tetrachloride (CCl4)-induced liver fibrosis were used as the animal model. Next, PZH treatment was given for 8 weeks. Afterward, the therapeutic effects of PZH were analyzed through a hepatic tissue structure by hematoxylin-eosin (H&E), Van Gieson (VG) staining, and transmission electron microscopy (TEM), activity of ALT and AST by enzyme-associated immunosorbent assay as well. Subsequently, mRNA and protein expression were examined by quantitative polymerase chain reaction (qPCR), Western blotting, and immunohistochemistry (IHC). Then, the cell vitality of PZH-treated HSC and the expression of key molecules prevailing to autophagy were studied in vitro. Meanwhile, SM16 (a novel small molecular inhibitor which inhibits TGFβ-induced Smad2 phosphorylation) was employed to confirm PZH’s effects on the proliferation and autophagy of HSC. Results: PZH pharmacologically exerted anti-hepatic fibrosis effects as demonstrated by protecting hepatocytes and improving hepatic function. The results revealed the reduced production of extracellular collagen by adjusting the balance of matrix metalloproteinase (MMP) 2, MMP9, and tissue inhibitor of matrix metalloproteinase 1 (TIMP1) in PZH-treated CCl4-induced liver fibrosis. Interestingly, PZH inhibited the activation of HSC by down-regulating TGF-β1 and phosphorylating Smad2. Furthermore, PZH down-regulated yeast Atg6 (Beclin-1) and microtubule-associated protein light chain 3 (LC3) toward suppressing HSC autophagy, and PZH exhibited similar effects to that of SM16. Conclusion: To conclude, PZH alleviated CCl4-induced liver fibrosis to reduce the production of extracellular collagen and inhibiting the activation of HSC. In addition, their pharmacological mechanisms related to autophagy and TGF-β1/Smad2 signaling pathways were revealed for the first time.
Collapse
Affiliation(s)
- Yuqin Zhang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Liping Hua
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Chunfeng Lin
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Mingzhou Yuan
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Wei Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Anand Raj D.
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore, Tamil Nadu, India
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - Carlos L. Cespedes-Acuna
- Plant Biochemistry and Phytochemical Ecology Lab, Basic Sciences Department University of Bio Bio, Chillan, Chile
| | - Shivraj Hariram Nile
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- *Correspondence: Shivraj Hariram Nile, ; Guohong Yan, ; Haiyin Zheng,
| | - Guohong Yan
- Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- *Correspondence: Shivraj Hariram Nile, ; Guohong Yan, ; Haiyin Zheng,
| | - Haiyin Zheng
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- *Correspondence: Shivraj Hariram Nile, ; Guohong Yan, ; Haiyin Zheng,
| |
Collapse
|
17
|
Zhang J, Shang L, Jiang W, Wu W. Shikonin induces apoptosis and autophagy via downregulation of pyrroline-5-carboxylate reductase1 in hepatocellular carcinoma cells. Bioengineered 2022; 13:7904-7918. [PMID: 35293266 PMCID: PMC9208523 DOI: 10.1080/21655979.2022.2052673] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Shikonin(SK) is a natural small molecule naphthoquinone compound, which has anti-cancer activity in various human malignant tumors. Pyrroline-5-carboxylate reductase 1(PYCR1) is involved in tumorigenesis and regulates various cellular processes, including growth, invasion, migration, and apoptosis. However, the effect of SK and PYCR1 on apoptosis and autophagy in hepatocellular carcinoma are unclear. Our goal is to determine the internal molecular mechanism of the interaction between SK and PYCR1 and its role in the occurrence and development of liver cancer. The CCK8 assay, wound healing assay, and transwell assays show that SK and siPYCR1(gene silence PYCR1) inhibited the malignant phenotype of HCC cells, including cell viability, colony formation, migration, and invasion, respectively. The flow cytometry assays and immunofluorescence show that SK and siPYCR1 activated apoptosis and autophagy, respectively. SK induces apoptosis and autophagy in a dose-dependent manner. In addition, HCC cells were transfected with small interference fragment PYCR1 siRNA to construct siPYCR1 and SK single treatment group and co-treatment group to verify the interaction between SK and PYCR1. The Western blot identified that PI3K/Akt/mTOR signal pathway protein expression was significantly downregulated in HCC cells treated with SK and siPYCR1 together. Collectively, SK may induce apoptosis and autophagy by reducing the expression of PYCR1 and suppressing PI3K/Akt/mTOR. Thus, SK may be a promising antineoplastic drug in Hepatocellular carcinoma (HCC). SK downregulating PYCR1 might supply a theoretical foundation for the potential therapeutic application in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Junli Zhang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Ling Shang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Wendi Jiang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Wenjuan Wu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| |
Collapse
|
18
|
Zhou JC, Wang JL, Ren HZ, Shi XL. Autophagy plays a double-edged sword role in liver diseases. J Physiol Biochem 2022; 78:9-17. [PMID: 34657993 PMCID: PMC8873123 DOI: 10.1007/s13105-021-00844-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
As a highly evolutionarily conserved process, autophagy can be found in all types of eukaryotic cells. Such a constitutive process maintains cellular homeostasis in a wide variety of cell types through the encapsulation of damaged proteins or organelles into double-membrane vesicles. Autophagy not only simply eliminates materials but also serves as a dynamic recycling system that produces new building blocks and energy for cellular renovation and homeostasis. Previous studies have primarily recognized the role of autophagy in the degradation of dysfunctional proteins and unwanted organelles. However, there are findings of autophagy in physiological and pathological processes. In hepatocytes, autophagy is not only essential for homeostatic functions but also implicated in some diseases, such as viral hepatitis, alcoholic hepatitis, and hepatic failure. In the present review, we summarized the molecular mechanisms of autophagy and its role in several liver diseases and put forward several new strategies for the treatment of liver disease.
Collapse
Affiliation(s)
- Jing-Chao Zhou
- Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Lin Wang
- Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hao-Zhen Ren
- Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Xiao-Lei Shi
- Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
19
|
Sun Q, Gong T, Liu M, Ren S, Yang H, Zeng S, Zhao H, Chen L, Ming T, Meng X, Xu H. Shikonin, a naphthalene ingredient: Therapeutic actions, pharmacokinetics, toxicology, clinical trials and pharmaceutical researches. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153805. [PMID: 34749177 DOI: 10.1016/j.phymed.2021.153805] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/15/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Shikonin is one of the major phytochemical components of Lithospermum erythrorhizon (Purple Cromwell), which is a type of medicinal herb broadly utilized in traditional Chinese medicine. It is well established that shikonin possesses remarkable therapeutic actions on various diseases, with the underlying mechanisms, pharmacokinetics and toxicological effects elusive. Also, the clinical trial and pharmaceutical study of shikonin remain to be comprehensively delineated. PURPOSE The present review aimed to systematically summarize the updated knowledge regarding the therapeutic actions, pharmacokinetics, toxicological effects, clinical trial and pharmaceutical study of shikonin. METHODS The information contained in this review article were retrieved from some authoritative databases including Web of Science, PubMed, Google scholar, Chinese National Knowledge Infrastructure (CNKI), Wanfang Database and so on, till August 2021. RESULTS Shikonin exerts multiple therapeutic efficacies, such as anti-inflammation, anti-cancer, cardiovascular protection, anti-microbiomes, analgesia, anti-obesity, brain protection, and so on, mainly by regulating the NF-κB, PI3K/Akt/MAPKs, Akt/mTOR, TGF-β, GSK3β, TLR4/Akt signaling pathways, NLRP3 inflammasome, reactive oxygen stress, Bax/Bcl-2, etc. In terms of pharmacokinetics, shikonin has an unfavorable oral bioavailability, 64.6% of the binding rate of plasma protein, and enhances some metabolic enzymes, particularly including cytochrome P450. In regard to the toxicological effects, shikonin may potentially cause nephrotoxicity and skin allergy. The above pharmacodynamics and pharmacokinetics of shikonin have been validated by few clinical trials. In addition, pharmaceutical innovation of shikonin with novel drug delivery system such as nanoparticles, liposomes, microemulsions, nanogel, cyclodextrin complexes, micelles and polymers are beneficial to the development of shikonin-based drugs. CONCLUSIONS Shikonin is a promising phytochemical for drug candidates. Extensive and intensive explorations on shikonin are warranted to expedite the utilization of shikonin-based drugs in the clinical setting.
Collapse
Affiliation(s)
- Qiang Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ting Gong
- Department of Ultrasound, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, Sichuan, China
| | - Maolun Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Han Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Sha Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hui Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
20
|
Cheng QN, Yang X, Wu JF, Ai WB, Ni YR. Interaction of non‑parenchymal hepatocytes in the process of hepatic fibrosis (Review). Mol Med Rep 2021; 23:364. [PMID: 33760176 PMCID: PMC7986015 DOI: 10.3892/mmr.2021.12003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatic fibrosis (HF) is the process of fibrous scar formation caused by chronic liver injury of different etiologies. Previous studies have hypothesized that the activation of hepatic stellate cells (HSCs) is the central process in HF. The interaction between HSCs and surrounding cells is also crucial. Additionally, hepatic sinusoids capillarization, inflammation, angiogenesis and fibrosis develop during HF. The process involves multiple cell types that are highly connected and work in unison to maintain the homeostasis of the hepatic microenvironment, which serves a key role in the initiation and progression of HF. The current review provides novel insight into the intercellular interaction among liver sinusoidal endothelial cells, HSCs and Kupffer cells, as well as the hepatic microenvironment in the development of HF.
Collapse
Affiliation(s)
- Qi-Ni Cheng
- Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Xue Yang
- Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Jiang-Feng Wu
- Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- The People's Hospital of China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Wen-Bing Ai
- The Yiling Hospital of Yichang, Yichang, Hubei 443100, P.R. China
| | - Yi-Ran Ni
- Medical College, China Three Gorges University, Yichang, Hubei 443002, P.R. China
- Institute of Organ Fibrosis and Targeted Drug Delivery, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| |
Collapse
|
21
|
Shu Y, Liu X, Huang H, Wen Q, Shu J. Research progress of natural compounds in anti-liver fibrosis by affecting autophagy of hepatic stellate cells. Mol Biol Rep 2021; 48:1915-1924. [PMID: 33609264 PMCID: PMC7925445 DOI: 10.1007/s11033-021-06171-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Chronic liver diseases caused by various pathogenesis are marked by inflammatory infiltration and wound healing reaction, while their normal regeneration ability is impaired. The unbalance between the generation and the degradation of extracellular matrix (ECM) leads to collagen accumulation and develops into liver fibrosis. Inflammation, oxidative stress, and autophagy interact closely in the pathogenesis of hepatic fibrosis. Reactive Oxygen Species (ROS) can not only stimulate Kupffer cells to release massive inflammatory factors, but induce autophagy. However, the latter may suppress inflammatory reaction by inhibiting proinflammatory complex formation directly, and removing damaged organelles or pathogenic microorganism indirectly. At present, effective anti-fibrosis drugs are still lacking. Previous studies have found various natural compounds enabled liver protection through anti-inflammatory, antioxidant, and other mechanisms. In recent years, autophagy, a vital life activity, has been found to be involved in the mechanism of liver fibrosis. As a new target, developing anti-liver fibrosis drugs that regulate the activity of autophagy is very promising. In this review, we summarize the latest studies about natural compounds in the treatment of liver fibrosis by regulating autophagy.
Collapse
Affiliation(s)
- Yongxiang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Xuyou Liu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Haifeng Huang
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Qi Wen
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Jianchang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| |
Collapse
|
22
|
Ji J, Yu Q, Dai W, Wu L, Feng J, Zheng Y, Li Y, Guo C. Apigenin Alleviates Liver Fibrosis by Inhibiting Hepatic Stellate Cell Activation and Autophagy via TGF- β1/Smad3 and p38/PPAR α Pathways. PPAR Res 2021; 2021:6651839. [PMID: 33574836 PMCID: PMC7861947 DOI: 10.1155/2021/6651839] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/10/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE The aim of this study is to confirm the hepatocellular protective functions of apigenin and the molecular mechanism on liver fibrosis in mice. METHODS Carbon tetrachloride (CCl4) and bile duct ligature (BDL) mouse fibrosis models were used to investigate the effects of apigenin on liver fibrosis. Sixty-six male C57 mice were randomly divided into eight groups, including the vehicle group, CCl4 group, CCl4+L-apigenin (20 mg/kg) group, CCl4+H-apigenin (40 mg/kg) group, sham group, BDL group, BDL+L-apigenin(20 mg/kg) group, and BDL+H-apigenin(40 mg/kg) group. Serum liver enzymes (ALT and AST), proteins associated with autophagy, and indicators linked with the TGF-β1/Smad3 and p38/PPARα pathways were detected using qRT-PCR, immunohistochemical staining, and western blotting. RESULTS Our findings confirmed that apigenin could decrease the levels of ALT and AST, suppress the generation of ECM, inhibit the activation of HSCs, regulate the balance of MMP2 and TIMP1, reduce the expression of autophagy-linked protein, and restrain the TGF-β1/Smad3 and p38/PPARα pathways. CONCLUSION Apigenin could alleviate liver fibrosis by inhibiting hepatic stellate cell activation and autophagy via TGF-β1/Smad3 and p38/PPARα pathways.
Collapse
Affiliation(s)
- Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
23
|
Puckett DL, Alquraishi M, Chowanadisai W, Bettaieb A. The Role of PKM2 in Metabolic Reprogramming: Insights into the Regulatory Roles of Non-Coding RNAs. Int J Mol Sci 2021; 22:1171. [PMID: 33503959 PMCID: PMC7865720 DOI: 10.3390/ijms22031171] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/17/2023] Open
Abstract
Pyruvate kinase is a key regulator in glycolysis through the conversion of phosphoenolpyruvate (PEP) into pyruvate. Pyruvate kinase exists in various isoforms that can exhibit diverse biological functions and outcomes. The pyruvate kinase isoenzyme type M2 (PKM2) controls cell progression and survival through the regulation of key signaling pathways. In cancer cells, the dimer form of PKM2 predominates and plays an integral role in cancer metabolism. This predominance of the inactive dimeric form promotes the accumulation of phosphometabolites, allowing cancer cells to engage in high levels of synthetic processing to enhance their proliferative capacity. PKM2 has been recognized for its role in regulating gene expression and transcription factors critical for health and disease. This role enables PKM2 to exert profound regulatory effects that promote cancer cell metabolism, proliferation, and migration. In addition to its role in cancer, PKM2 regulates aspects essential to cellular homeostasis in non-cancer tissues and, in some cases, promotes tissue-specific pathways in health and diseases. In pursuit of understanding the diverse tissue-specific roles of PKM2, investigations targeting tissues such as the kidney, liver, adipose, and pancreas have been conducted. Findings from these studies enhance our understanding of PKM2 functions in various diseases beyond cancer. Therefore, there is substantial interest in PKM2 modulation as a potential therapeutic target for the treatment of multiple conditions. Indeed, a vast plethora of research has focused on identifying therapeutic strategies for targeting PKM2. Recently, targeting PKM2 through its regulatory microRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) has gathered increasing interest. Thus, the goal of this review is to highlight recent advancements in PKM2 research, with a focus on PKM2 regulatory microRNAs and lncRNAs and their subsequent physiological significance.
Collapse
Affiliation(s)
- Dexter L. Puckett
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Mohammed Alquraishi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Winyoo Chowanadisai
- Department of Nutrition, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| |
Collapse
|
24
|
Yu Q, Cheng P, Wu J, Guo C. PPARγ/NF-κB and TGF-β1/Smad pathway are involved in the anti-fibrotic effects of levo-tetrahydropalmatine on liver fibrosis. J Cell Mol Med 2021; 25:1645-1660. [PMID: 33438347 PMCID: PMC7875896 DOI: 10.1111/jcmm.16267] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Liver fibrosis is a necessary stage in the development of chronic liver diseases to liver cirrhosis. This study aims to investigate the anti‐fibrotic effects of levo‐tetrahydropalmatine (L‐THP) on hepatic fibrosis in mice and cell models and its underlying mechanisms. Two mouse hepatic fibrosis models were generated in male C57 mice by intraperitoneal injection of carbon tetrachloride (CCl4) for 2 months and bile duct ligation (BDL) for 14 days. Levo‐tetrahydropalmatine was administered orally at doses of 20 and 40 mg/kg. An activated LX2 cell model induced by TGF‐β1 was also generated. The results showed that levo‐tetrahydropalmatine alleviated liver fibrosis by inhibiting the formation of extracellular matrix (ECM) and regulating the balance between TIMP1 and MMP2 in the two mice liver fibrosis models and cell model. Levo‐tetrahydropalmatine inhibited activation and autophagy of hepatic stellate cells (HSCs) by modulating PPARγ/NF‐κB and TGF‐β1/Smad pathway in vivo and in vitro. In conclusion, levo‐tetrahydropalmatine attenuated liver fibrosis by inhibiting ECM deposition and HSCs autophagy via modulation of PPARγ/NF‐κB and TGF‐β1/Smad pathway.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping Cheng
- Department of Gerontology, Shanghai Minhang District Central Hospital, Shanghai, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Bergenin Attenuates Hepatic Fibrosis by Regulating Autophagy Mediated by the PPAR- γ/TGF- β Pathway. PPAR Res 2020; 2020:6694214. [PMID: 33488687 PMCID: PMC7790590 DOI: 10.1155/2020/6694214] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/06/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is a pathological process involving diffuse extracellular matrix (ECM) deposition in the liver. It is typical of many chronic liver diseases, including cirrhosis, and effective drugs are needed. In this study, we explored the protective effect of bergenin on liver fibrosis induced by carbon tetrachloride and bile duct ligation. A variety of molecular biological methods (qRT-PCR, western blotting, and immunohistochemistry) were employed to confirm the increased degree of hepatocyte injury and ECM formation in the disease model, consistent with autophagy and activation of the TGF-β pathway. Bergenin activated PPAR-γ and inhibited TGF-β and autophagy and decreased liver fibrosis by inhibiting hepatocyte necrosis and ECM formation in a dose-dependent manner. The results suggest that bergenin may be a promising drug candidate for the treatment of liver fibrosis.
Collapse
|
26
|
Shen XB, Wang Y, Han XZ, Sheng LQ, Wu FF, Liu X. Design, synthesis and anticancer activity of naphthoquinone derivatives. J Enzyme Inhib Med Chem 2020; 35:773-785. [PMID: 32200656 PMCID: PMC7144209 DOI: 10.1080/14756366.2020.1740693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Basis on molecular docking and pharmacophore analysis of naphthoquinone moiety, a total of 23 compounds were designed and synthesised. With the help of reverse targets searching, anti-cancer activity was preliminarily evaluated, most of them are effective against some tumour cells, especially compound 12: 1-(5,8-dihydroxy-1,4-dioxo-1,4-dihydronaphthalen-2-yl)-4-methylpent-3-en-1-yl-4-oxo-4-((4-phenoxyphenyl)amino) butanoate whose IC50 against SGC-7901 was 4.1 ± 2.6 μM. Meanwhile the anticancer mechanism of compound 12 had been investigated by AnnexinV/PI staining, immunofluorescence, Western blot assay and molecular docking. The results indicated that this compound might induce cell apoptosis and cell autophagy through regulating the PI3K signal pathway.
Collapse
Affiliation(s)
- Xiao-Bao Shen
- School of Pharmacy, Anhui Medical University, Hefei, PR China.,Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Yang Wang
- School of Pharmacy, Anhui Medical University, Hefei, PR China
| | - Xuan-Zhen Han
- Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Liang-Quan Sheng
- Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Fu-Fang Wu
- School of Pharmacy, Anhui Medical University, Hefei, PR China.,Engineering Research Center of Biomass Conversion and Pollution Prevention of Anhui Educational Institutions, Fuyang Normal University, Fuyang, PR China
| | - Xinhua Liu
- School of Pharmacy, Anhui Medical University, Hefei, PR China
| |
Collapse
|
27
|
Wang Y, Zhu F, Zhang Y, Chen C, Lai Y, Sun J, Chen S, Qiu P, Gao J, Deng G. Shikonin suppresses trophoblast cell growth via regulation of GLI1, and p62 mediated caspase 8 activation. Reprod Toxicol 2020; 95:104-112. [PMID: 32461113 DOI: 10.1016/j.reprotox.2020.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/10/2020] [Accepted: 05/18/2020] [Indexed: 10/24/2022]
Abstract
Unruptured ectopic pregnancy (UEP) is a common cause of morbidity and, occasionally, of mortality in women of reproductive age. Pharmacological intervention is a common therapeutic approach for early-stage UEP. Herein, we investigated the cytotoxic effect and novel mechanism of shikonin, a natural naphthoquinone pigment purified from Lithospermum erythrorhizon, in human trophoblast cells. These data demonstrated that shikonin suppressed proliferation and induced apoptosis in a time-dependent manner in HTR-8/SVneo cells. Shikonin blocked autophagic flux and promoted p62 interaction with caspase 8, resulting in caspase 8 activation. Moreover, shikonin suppressed GLI1 expression, and GLI1 overexpression attenuated shikonin-induced cell apoptosis. Although silencing GLI1 slightly promoted cell apoptosis, p62 overexpression enhanced GLI1 silencing-induced cell apoptosis by activating caspase 8. Furthermore, rapamycin increased shikonin-induced cell apoptosis in HTR-8/SVneo cells, whereas 3-MA attenuated the cytotoxic effect of shikonin. In conclusion, shikonin suppressed trophoblast cell growth by silencing GLI1 and increasing p62 co-mediated activation of caspase 8, which suggested a potential novel therapeutic target for UEP.
Collapse
Affiliation(s)
- Yanxi Wang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Fangfang Zhu
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yingxuan Zhang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chunlin Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yuling Lai
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jianhua Sun
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Si Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Pin Qiu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jie Gao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Gaopi Deng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
28
|
Xu S, Mao Y, Wu J, Feng J, Li J, Wu L, Yu Q, Zhou Y, Zhang J, Chen J, Ji J, Chen K, Wang F, Dai W, Fan X, Guo C. TGF-β/Smad and JAK/STAT pathways are involved in the anti-fibrotic effects of propylene glycol alginate sodium sulphate on hepatic fibrosis. J Cell Mol Med 2020; 24:5224-5237. [PMID: 32233073 PMCID: PMC7205790 DOI: 10.1111/jcmm.15175] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 01/14/2020] [Accepted: 03/01/2020] [Indexed: 12/25/2022] Open
Abstract
Liver fibrosis, a consequence of unhealthy modern lifestyles, has a growing impact on human health, particularly in developed countries. Here, we have explored the anti-fibrotic effects of propylene glycol alginate sodium sulphate (PSS), a natural extract from brown algae, in fibrotic mice and cell models. Thus, we established bile duct ligature and carbon tetrachloride mouse models and LX-2 cell models with or without PSS treatment. Liver pathological sections and the relevant indicators in serum and liver tissues were examined. PSS prevented hepatic injury and fibrosis to a significant extent, and induced up-regulation of matrix metalloproteinase-2 and down-regulation of tissue inhibitor of metalloproteinase-1 through suppressing the transforming growth factor β1 (TGF-β1)/Smad pathway. PSS additionally exerted an anti-autophagy effect through suppressing the Janus kinase (JAK) 2/transducer and activator of transcription 3 (STAT3) pathway. In conclusion, PSS prevents hepatic fibrosis by suppressing inflammation, promoting extracellular matrix (ECM) decomposition and inactivating hepatic stellate cells through mechanisms involving the TGF-β1/Smad2/3 and JAK2/STAT3 pathways in vivo and in vitro.
Collapse
Affiliation(s)
- Shizan Xu
- Department of GastroenterologyPutuo People's HospitalTongji University School of MedicineShanghaiChina
- Department of GastroenterologyJinshan Hospital of Fudan UniversityShanghaiChina
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Yuqing Mao
- Department of GerontologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianye Wu
- Department of GastroenterologyPutuo People's HospitalTongji University School of MedicineShanghaiChina
| | - Jiao Feng
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
| | - Jingjing Li
- Department of GastroenterologyPutuo People's HospitalTongji University School of MedicineShanghaiChina
| | - Liwei Wu
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
| | - Qiang Yu
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Yuting Zhou
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jie Zhang
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jiaojiao Chen
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jie Ji
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
| | - Kan Chen
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
| | - Fan Wang
- Department of OncologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weiqi Dai
- Department of GastroenterologyPutuo People's HospitalTongji University School of MedicineShanghaiChina
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
- Department of GastroenterologyZhongshan Hospital of Fudan UniversityShanghaiChina
- Shanghai Institute of Liver DiseasesZhongshan Hospital of Fudan UniversityShanghaiChina
- Shanghai Tongren HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaoming Fan
- Department of GastroenterologyJinshan Hospital of Fudan UniversityShanghaiChina
| | - Chuanyong Guo
- Department of GastroenterologyPutuo People's HospitalTongji University School of MedicineShanghaiChina
- Department of GastroenterologyShanghai Tenth People’s HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
29
|
Gwon SY, Ahn J, Jung CH, Moon B, Ha TY. Shikonin Attenuates Hepatic Steatosis by Enhancing Beta Oxidation and Energy Expenditure via AMPK Activation. Nutrients 2020; 12:nu12041133. [PMID: 32316687 PMCID: PMC7230385 DOI: 10.3390/nu12041133] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/22/2022] Open
Abstract
Shikonin, a natural plant pigment, is known to have anti-obesity activity and to improve insulin sensitivity. This study aimed to examine the effect of shikonin on hepatic steatosis, focusing on the AMP-activated protein kinase (AMPK) and energy expenditure in Hepa 1-6 cells and in high-fat fed mice. Shikonin increased AMPK phosphorylation in a dose- and time-dependent manner, and inhibition of AMPK with compound C inhibited this activation. In an oleic acid-induced steatosis model in hepatocytes, shikonin suppressed oleic acid-induced lipid accumulation, increased AMPK phosphorylation, suppressed the expression of lipogenic genes, and stimulated fatty acid oxidation-related genes. Shikonin administration for four weeks decreased body weight gain and the accumulation of lipid droplets in the liver of high-fat fed mice. Furthermore, shikonin promoted energy expenditure by activating fatty acid oxidation. In addition, shikonin increased the expression of PPARγ coactivator-1α (PGC-1α), carnitine palmitoyltransferase-1 (CPT1) and other mitochondrial function-related genes. These results suggest that shikonin attenuated a high fat diet-induced nonalcoholic fatty liver disease by stimulating fatty acid oxidation and energy expenditure via AMPK activation.
Collapse
Affiliation(s)
- So Young Gwon
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.G.); (J.A.); (C.H.J.)
- Department of Law Policy Research, National Food Safety Information Service, Seoul 110-750, Korea
| | - Jiyun Ahn
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.G.); (J.A.); (C.H.J.)
- Division of Food Biotechnology, University of Science & Technology, Daejeon 305-350, Korea
| | - Chang Hwa Jung
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.G.); (J.A.); (C.H.J.)
- Division of Food Biotechnology, University of Science & Technology, Daejeon 305-350, Korea
| | - BoKyung Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea;
| | - Tae-Youl Ha
- Division of Food Functionality Research, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.G.); (J.A.); (C.H.J.)
- Division of Food Biotechnology, University of Science & Technology, Daejeon 305-350, Korea
- Correspondence: ; Tel.: +82-632299054; Fax: +82-632299225
| |
Collapse
|
30
|
Ma L, Wang X, Li W, Qu F, Liu Y, Lu J, Su G, Zhao Y. Conjugation of Ginsenoside with Dietary Amino Acids: A Promising Strategy To Suppress Cell Proliferation and Induce Apoptosis in Activated Hepatic Stellate Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:10245-10255. [PMID: 31389238 DOI: 10.1021/acs.jafc.9b03305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ginseng has been widely used as a functional food in the world because of its well-defined health benefits. Previous studies have confirmed that AD-1, a new ginsenoside derived from ginseng, can ameliorate thioacetamide-induced liver injury and fibrosis in mice. Simultaneously, amino acid supplementation is getting more attention as an important adjuvant therapy in the improvement of hepatopathy. The aim of this study was to conjugate AD-1 with several selected amino acids and investigate the cytotoxicity of the obtained conjugates in activated t-HSC/Cl-6 cells and normal human liver cells (LO2). Structure-activity relationships of conjugates and underlying mechanisms of the effect are also explored. The results indicated that conjugate 7c remarkably inhibited cell proliferation in activated t-HSC/Cl-6 cells (IC50 = 3.8 ± 0.4 μM) and appeared to be nontoxic to LO2. Besides, conjugate 7c had a relatively good plasma stability. Further study demonstrated that inducing S-phase arrest and activation of mitochondrial-mediated apoptosis were included in the mechanisms underlying the efficiency of conjugate 7c. These findings provided further insight into designing functional foods (ginsenoside and amino acid) for the application in prevention or improvement of liver fibrosis.
Collapse
|
31
|
Yang X, Wang H, Tu Y, Li Y, Zou Y, Li G, Wang L, Zhong X. WNT1-inducible signaling protein-1 mediates TGF-β1-induced renal fibrosis in tubular epithelial cells and unilateral ureteral obstruction mouse models via autophagy. J Cell Physiol 2019; 235:2009-2022. [PMID: 31512238 DOI: 10.1002/jcp.29187] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022]
Abstract
Renal fibrosis is a common pathway for the progression of all chronic kidney diseases to end-stage kidney disease. Studies show that WNT1-inducible signaling pathway protein-1 (WISP-1) is involved in the fibrosis of various organs. The aim of the study was to explore the functional role and potential mechanism of WISP-1 in renal fibrosis. We observed that overexpression of WISP-1 in rat tubular epithelial cells (TECs) enhanced transforming growth factor-β1 (TGF-β1)-induced production of fibrotic markers, including collagen I (Col I), fibronectin (FN) and TGF-β1, while inhibition of WISP-1 suppressed such production. In vivo, the messenger RNA and protein levels of Col I, FN, and α-smooth muscle actin were significantly inhibited after anti-WISP-1 antibody treatment for 7 days in unilateral ureteral obstruction mouse models. Moreover, blockade of WISP-1 by anti-WISP-1 antibody significantly reduced autophagy-related markers, including anti-microtubule-associated protein-1 light chain 3 (LC3) and beclin 1, while increasing sequestosome 1. In addition, overexpression of WISP-1 in TECs increased autophagy as evidenced by greater numbers of GFP-LC3 puncta and increased expression of LC3 and beclin 1 in response to TGF-β1. In contrast, knockdown of WISP-1 by small interfering RNA decreased the number of GFP-LC3 puncta and the expression of LC3 and beclin 1 in TGF-β1-treated TECs. Collectively, these data suggest that WISP-1, as a profibrotic protein, may mediate renal fibrosis by inducing autophagy in both obstructive nephropathy and TGF-β1-treated TECs. WISP-1 may serve as an effective therapeutic target for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Xue Yang
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Nephrology, Du Jiang Yan Medical Center, Chengdu, China
| | - Huan Wang
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yueju Tu
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Li
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yurong Zou
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guisen Li
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Wang
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiang Zhong
- Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
32
|
Wang HY, Li C, Liu WH, Deng FM, Ma Y, Guo LN, Kong DH, Hu KA, Liu Q, Wu J, Sun J, Liu YL. Autophagy inhibition via Becn1 downregulation improves the mesenchymal stem cells antifibrotic potential in experimental liver fibrosis. J Cell Physiol 2019; 235:2722-2737. [PMID: 31508820 PMCID: PMC6916329 DOI: 10.1002/jcp.29176] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023]
Abstract
Liver fibrosis (LF) is the result of a vicious cycle between inflammation-induced chronic hepatocyte injury and persistent activation of hepatic stellate cells (HSCs). Mesenchymal stem cell (MSC)-based therapy may represent a potential remedy for treatment of LF. However, the fate of transplanted MSCs in LF remains largely unknown. In the present study, the fate and antifibrotic effect of MSCs were explored in a LF model induced by CCl4 in mouse. Additionally, MSCs were stimulated in vitro with LF-associated factors, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), and transforming growth factor-β1 (TGF-β1), to mimic the LF microenvironment. We unveiled that MSCs exhibited autophagy in response to the LF microenvironment through Becn1 upregulation both in vivo and in vitro. However, autophagy suppression induced by Becn1 knockdown in MSCs resulted in enhanced antifibrotic effects on LF. The improved antifibrotic potential of MSCs may be attributable to their inhibitory effects on T lymphocyte infiltration, HSCs proliferation, as well as production of TNF-α, IFN-γ, and TGF-β1, which may be partially mediated by elevated paracrine secretion of PTGS2/PGE2 . Thus, autophagy manipulation in MSCs may be a novel strategy to promote their antifibrotic efficacy.
Collapse
Affiliation(s)
- Hang Yu Wang
- Key Laboratory of Xingjiang Phytomedicine Resources and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Can Li
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China.,Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wei Hua Liu
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Feng Mei Deng
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China.,Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yan Ma
- Department of Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Li Na Guo
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| | - De Hua Kong
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Kang An Hu
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Qin Liu
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jiang Wu
- Deep-Underground Medicine Laboratory, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Sun
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China.,Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yi Lun Liu
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China.,Department of Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
33
|
Feng J, Wang C, Liu T, Li J, Wu L, Yu Q, Li S, Zhou Y, Zhang J, Chen J, Ji J, Chen K, Mao Y, Wang F, Dai W, Fan X, Wu J, Guo C. Procyanidin B2 inhibits the activation of hepatic stellate cells and angiogenesis via the Hedgehog pathway during liver fibrosis. J Cell Mol Med 2019; 23:6479-6493. [PMID: 31328391 PMCID: PMC6714206 DOI: 10.1111/jcmm.14543] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/08/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Liver fibrosis is a wound-healing process of liver featured by the over-deposition of extracellular matrix (ECM) and angiogenesis. However, the effective treatment is lacking. Procyanidin B2 (PB2) is a flavonoid extract abundant in grape seeds with anti-oxidant, anti-inflammatory and anti-cancer properties. The present study aimed to determine effects of PB2 on liver fibrosis. METHOD The CCl4-induced mouse liver fibrosis model and a human hepatic stellate cell (HSC) line (LX2 cells) were used to study the activation, ECM production and angiogenesis of HSCs through Western blotting analysis, immunohistochemistry, immunofluorescence staining, flow cytometry and tubulogenesis assay. A Hedgehog (Hh) pathway inhibitor (cyclopamine) and Smoothened agonist (SAG) were used to investigate the role of PB2 on Hh pathway. RESULTS The results showed that PB2 could inhibit the proliferation and induce apoptosis of HSCs. PB2 could also down-regulate the expressions of VEGF-A, HIF-1α, α-SMA, Col-1 and TGF-β1 of HSCs in vivo and in vitro. The application of SAG and cyclopamine proved that PB2 targets on Hh pathway. CONCLUSIONS PB2 inhibited the Hh pathway to suppress the activation, ECM production and angiogenesis of HSCs, therefore reverses the progression of liver fibrosis in vivo and in vitro.
Collapse
Affiliation(s)
- Jiao Feng
- Department of Gastroenterology, Putuo People's HospitalTongji University School of MedicineShanghaiChina
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Chengfen Wang
- Department of Gastroenterology, Putuo People's HospitalTongji University School of MedicineShanghaiChina
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's HospitalTongji University School of MedicineShanghaiChina
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Yuting Zhou
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jie Zhang
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jiaojiao Chen
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Shanghai Tenth HospitalSchool of Clinical Medicine of Nanjing Medical UniversityShanghaiChina
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Yuqing Mao
- Department of Gerontology, Shanghai General HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Fan Wang
- Department of Oncology, Shanghai General HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's HospitalTongji University School of MedicineShanghaiChina
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Department of GastroenterologyZhongshan Hospital of Fudan UniversityShanghaiChina
- Shanghai Institute of Liver DiseasesZhongshan Hospital of Fudan UniversityShanghaiChina
| | - Xiaoming Fan
- Department of GastroenterologyJinshan Hospital of Fudan UniversityJinshan, ShanghaiChina
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's HospitalTongji University School of MedicineShanghaiChina
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
34
|
Locostatin Alleviates Liver Fibrosis Induced by Carbon Tetrachloride in Mice. Dig Dis Sci 2019; 64:2570-2580. [PMID: 30874989 DOI: 10.1007/s10620-019-05588-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Liver fibrosis is featured with excessive deposition of extracellular matrix and fibrous connective tissue hyperplasia. The specific inhibitor of Raf-1 kinase inhibitor protein, locostatin, inhibits the migration of hepatic stellate cells. In this study, we investigated the effect of locostatin on liver fibrosis and its underlying mechanism. METHODS Carbon tetrachloride (CCl4) was used to induce liver fibrosis in mice, and locostatin was injected intraperitoneally. Liver fibrosis was assessed by Masson and Sirius red staining, hydroxyproline (HYP) assay, and collagen percentage area. Collagen I, collagen III, and α-SMA were detected by RT-PCR and western blot. The levels of MMP-13, MMP-2, TIMP-1, and TIMP-2 were estimated by ELISA. Liver inflammation was evaluated by HE staining and immunohistochemistry; liver myeloperoxidase (MPO), superoxide dismutase, and malondialdehyde were measured by ELISA; and cytokines were by Mouse Cytokine Array Q4000. RESULTS Compared to the CCl4 group, HYP (208.56 ± 6.12) µg/g, percentage of total collagen at overall region (1.91 ± 0.13), MMP-13/TIMP-1 (0.19 ± 0.01), MPO (1.45 ± 0.04) U/g, TGF-β (2652 ± 91.20), PDGF-AA (3897 ± 290.69), and E-selectin (1569 ± 66.48) in the liver tissues were decreased significantly in the locostatin-treated group. CONCLUSIONS Locostatin mitigated liver fibrosis and inflammation induced by CCl4. The mechanism is via inhibition inflammatory cytokines, TGF-β, PDGF-AA, and E-selectin.
Collapse
|
35
|
Isorhamnetin Inhibits Liver Fibrosis by Reducing Autophagy and Inhibiting Extracellular Matrix Formation via the TGF- β1/Smad3 and TGF- β1/p38 MAPK Pathways. Mediators Inflamm 2019; 2019:6175091. [PMID: 31467486 PMCID: PMC6701280 DOI: 10.1155/2019/6175091] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/25/2019] [Accepted: 06/16/2019] [Indexed: 02/07/2023] Open
Abstract
Objective Liver fibrosis is a consequence of wound-healing responses to chronic liver insult and may progress to liver cirrhosis if not controlled. This study investigated the protection against liver fibrosis by isorhamnetin. Methods Mouse models of hepatic fibrosis were established by intraperitoneal injection of carbon tetrachloride (CCl4) or bile duct ligation (BDL). Isorhamnetin 10 or 30 mg/kg was administered by gavage 5 days per week for 8 weeks in the CCl4 model and for 2 weeks in the BDL model. Protein and mRNA expressions were assayed by western blotting, immunohistochemistry, and quantitative real-time polymerase chain reaction. Results Isorhamnetin significantly inhibited liver fibrosis in both models, inhibiting hepatic stellate cell (HSC) activation, extracellular matrix (ECM) deposition, and autophagy. The effects were associated with downregulation of transforming growth factor β1 (TGF-β1) mediation of Smad3 and p38 mitogen-activated protein kinase (MAPK) signaling pathways. Conclusion Isorhamnetin protected against liver fibrosis by reducing ECM formation and autophagy via inhibition of TGF-β1-mediated Smad3 and p38 MAPK signaling pathways.
Collapse
|
36
|
Active Constituent in the Ethyl Acetate Extract Fraction of Terminalia bellirica Fruit Exhibits Antioxidation, Antifibrosis, and Proapoptosis Capabilities In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5176090. [PMID: 31210842 PMCID: PMC6532289 DOI: 10.1155/2019/5176090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/14/2019] [Accepted: 04/21/2019] [Indexed: 02/06/2023]
Abstract
Terminalia bellirica (Gaertn.) Roxb. fruit (TBF) is a widely planted traditional medicinal herb in Tibet. We aimed to determine the most active substance-enriched extract by comparing the in vitro antioxidant activities of different extract fractions of TBF that were subsequently extracted by petroleum ether, chloroform, ethyl acetate, and n-butanol after initial extraction by 95% ethanol. The main compounds of the ethyl acetate extract fraction (EF) were analyzed via HPLC-MS. Gallic acid (GA) was obtained from EF to determine in vitro antifibrotic activity based on the traditional usage of TBF. After HSC-T6 cells were incubated with GA, extracellular secreted levels of fibrosis-associated cytokines, such as collagen I, collagen III, TGF-β1, and hydroxyproline, were estimated by ELISA. Gene and protein expressions of PDGFR, CTGF, NF-κB, MMP-2, TIMP-1, TIMP-2, α-SMA, and the Bcl-2/Bax family were determined by quantitative PCR and western blot. The proapoptotic effect of GA was further investigated by annexin V-PI and TUNEL staining. These results indicate that EF has prominent in vitro antioxidant activity among four extract fractions, and its main component, GA, manifests antifibrosis activity and its potential mechanism of action includes inhibition of cytokine secretion and collagen synthesis, as well as proapoptosis of HSCs.
Collapse
|
37
|
Autophagy promotes hepatic differentiation of hepatic progenitor cells by regulating the Wnt/β-catenin signaling pathway. J Mol Histol 2019; 50:75-90. [PMID: 30604254 PMCID: PMC6323068 DOI: 10.1007/s10735-018-9808-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022]
Abstract
Hepatic progenitor cells (HPCs) can be activated when the liver suffers persistent and severe damage and can differentiate into hepatocytes to maintain liver regeneration and homeostasis. However, the molecular mechanism underlying the hepatic differentiation of HPCs is unclear. Therefore, in this study, we aimed to investigate the roles of autophagy and the Wnt/β-catenin signaling pathway during hepatic differentiation of HPCs in vivo and in vitro. First, immunohistochemistry, immunofluorescence and electron microscopy showed that Atg5 and β-catenin were highly expressed in human fibrotic liver and mouse liver injury induced by feeding a 50% choline-deficient diet plus 0.15% ethionine solution in drinking water (CDE diet) for 21 days; in addition, these factors were expressed in CK19-positive HPCs. Second, Western blotting and immunofluorescence confirmed that CK19-positive HPCs incubated in differentiation medium for 7 days can differentiate into hepatocytes and that differentiated HPCs were able to take up ICG and secrete albumin and urea. Further investigation via Western blotting, immunofluorescence and electron microscopy revealed autophagy and the Wnt/β-catenin pathway to be activated during hepatic differentiation of HPCs. Next, we found that inhibiting autophagy by downregulating Atg5 gene expression impaired hepatic differentiation of HPCs and inhibited activation of the Wnt/β-catenin pathway, which was rescued by overexpression of the β-catenin gene. Moreover, downregulating β-catenin gene expression without inhibiting autophagy still impeded the differentiation of HPCs. Finally, coimmunoprecipitation demonstrated that P62 forms a complex with phosphorylated glycogen synthase kinase 3 beta (pGSK3β). Third, in mouse CDE-induced liver injury, immunohistochemistry and immunofluorescence confirmed that downregulating Atg5 gene expression inhibited autophagy, thus impeding hepatic differentiation of HPCs and inhibiting activation of the Wnt/β-catenin pathway. As observed in vitro, overexpression of β-catenin rescued this phenomenon caused by autophagy inhibition, though decreasing β-catenin levels without autophagy inhibition still impeded HPC differentiation. We also found that HPCs differentiated into hepatocytes in human fibrotic liver tissue. Collectively, these results demonstrate that autophagy promotes HPC differentiation by regulating Wnt/β-catenin signaling. Our results are the first to identify a role for autophagy in promoting the hepatic differentiation of HPCs.
Collapse
|
38
|
Wang XY, Liu WL. Mechanism of autophagy in liver fibrosis. Shijie Huaren Xiaohua Zazhi 2018; 26:1415-1422. [DOI: 10.11569/wcjd.v26.i23.1415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autophagy is an evolutionarily conserved lysosome-dependent catabolic process which degrades cell components, including proteins and lipids, in order to recycle substrates to exert optimally and adapt to tough circumstances. It is an important mechanism for the body to maintain the homeostasis of the internal environment. Liver fibrosis refers to the excessive proliferation and abnormal deposition of extracellular matrix components in the liver tissue, resulting in pathological changes in liver structure and function abnormalities, which is seen in chronic liver diseases of many different causes. In this article, we summarizes the role of autophagy in hepatic fibrosis as well as the relevant signaling pathways to reveal the mechanism of autophagy in hepatic fibrosis.
Collapse
Affiliation(s)
- Xin-Yan Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wen-Lan Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|