1
|
Hao LS, Ji JX, Jiang MY, Song J, Chen PP, Zhan ZY, Miao XJ, Gao YY, Wang W, Liu T. Effects of changes in SHP2 expression on liver fibrosis by influencing the apoptosis of hepatic stellate cells. APMIS 2025; 133:e13487. [PMID: 39500724 DOI: 10.1111/apm.13487] [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: 01/05/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024]
Abstract
Accumulating research has revealed that src-homology domain 2-containing protein tyrosine phosphatase-2 (SHP2), an oncogenic protein tyrosine phosphatase, is associated with liver fibrosis. Currently, it is still unclear whether SHP2 affects liver fibrosis by influencing the apoptosis of hepatic stellate cells (HSC). In present study, we investigate effects of SHP2 expression changes on liver fibrosis, with special emphasis on the apoptosis of HSC. Using adenovirus vector, wild-type SHP2 gene and short hairpin RNA targeting SHP2 were introduced into rats with liver fibrosis and LX-2 cells in vitro. The expressions of type I and III collagen, pathological and functional changes, collagen deposition in rat liver and apoptosis of LX-2 cells were detected by immunohistochemical and HE staining, automated biochemical analyzer, Masson trichrome staining, and TUNEL. This study showed that overexpression of SHP2 exacerbated dysfunction, inflammatory damage, collagen deposition and increased expression of type I and III collagen in rat liver reduced apoptosis of LX-2 cells. On the contrary, low expression of SHP2 alleviated the aforementioned detection indicators of rats and promoted apoptosis of LX-2 cells. In conclusion, the downregulation of SHP2 expression alleviates liver fibrosis by inducing the apoptosis of HSC, while overexpressed SHP2 exacerbates liver fibrosis by inhibiting the apoptosis of HSC.
Collapse
Affiliation(s)
- Li-Sen Hao
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Jing-Xiu Ji
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Mei-Yu Jiang
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Jie Song
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Pan-Pan Chen
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Zong-Yuan Zhan
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Xiao-Jia Miao
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Ying-Ying Gao
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Wei Wang
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Tian Liu
- Department of Gastroenterology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei Province, China
| |
Collapse
|
2
|
Sun Z, Chen G. Impact of heterogeneity in liver matrix and intrahepatic cells on the progression of hepatic fibrosis. Tissue Cell 2024; 91:102559. [PMID: 39293139 DOI: 10.1016/j.tice.2024.102559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/05/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
Liver fibrosis is a disease with a high prevalence worldwide. The development of hepatic fibrosis results from a combination of factors within the liver, such as extracellular matrix (ECM) deposition, hepatic stellate cells (HSCs) activation, collagen cross-linking, and inflammatory response. Heterogeneity in fibrotic liver is the result of a combination of heterogeneity in the intrahepatic microenvironment as well as heterogeneous expression of fibrosis-associated enzymes and cells, complicating the study of the mechanisms underlying the progression of liver fibrosis. The role of this heterogeneity on the crosstalk between cells and matrix and on the fibrotic process is worth exploring. In this paper, we will describe the phenomenon and mechanism of heterogeneity of liver matrix and intrahepatic cells in the process of hepatic fibrosis and discuss the crosstalk between heterogeneous factors on the development of fibrosis. The elucidation of heterogeneity is important for a deeper understanding of the pathological mechanisms of liver fibrosis as well as for clinical diagnosis and targeted therapies.
Collapse
Affiliation(s)
- Zhongtao Sun
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| |
Collapse
|
3
|
Trillos-Almanza MC, Aguilar MM, Buist-Homan M, Bomer N, Gomez KA, de Meijer VE, van Vilsteren FGI, Blokzijl H, Moshage H. Branched-chain amino acids and their metabolites decrease human and rat hepatic stellate cell activation. Mol Biol Rep 2024; 51:1116. [PMID: 39495311 PMCID: PMC11534903 DOI: 10.1007/s11033-024-10027-4] [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: 08/19/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND End-stage liver diseases (ESLDs) are a significant global health challenge due to their high prevalence and severe health impacts. Despite the severe outcomes associated with ESLDs, therapeutic options remain limited. Targeting the activation of hepatic stellate cells (HSCs), key drivers of extracellular matrix accumulation during liver injury presents a novel therapeutic approach. In ESLDs patients, branched-chain amino acids (BCAAs, leucine, isoleucine and valine) levels are decreased, and supplementation has been proposed to attenuate liver fibrosis and improve regeneration. However, their effects on HSCs require further investigation. OBJECTIVE To evaluate the efficacy of BCAAs and their metabolites, branched-chain α-keto acids (BCKAs), in modulating HSCs activation in human and rat models. METHODS Primary HSCs from rats and cirrhotic and non-cirrhotic human livers, were cultured and treated with BCAAs or BCKAs to assess their effects on both preventing (from day 1 of isolation) and reversing (from day 7 of isolation) HSCs activation. RESULTS In rat HSCs, leucine and BCKAs significantly reduced fibrotic markers and cell proliferation. In human HSCs, the metabolite of isoleucine decreased cell proliferation around 85% and increased the expression of branched-chain ketoacid dehydrogenase. The other metabolites also showed antifibrotic effects in HSCs from non-cirrhotic human livers. CONCLUSION BCAAs and their respective metabolites inhibit HSC activation with species-specific responses. Further research is needed to understand how BCAAs influence liver fibrogenesis. BCKAs supplementation could be a strategic approach for managing ESLDs, considering the nutritional status and amino acid profiles of patients.
Collapse
Affiliation(s)
- Maria Camila Trillos-Almanza
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| | - Magnolia Martinez Aguilar
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Karla Arevalo Gomez
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Vincent E de Meijer
- Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Frederike G I van Vilsteren
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
4
|
Shi M, Guo Y, Xu J, Yan L, Li X, Liu R, Feng Y, Zhang Y, Zhao Y, Zhang C, Du K, Li M, Zhang Y, Zhang J, Li Z, Ren D, Liu P. Gaudichaudione H ameliorates liver fibrosis and inflammation by targeting NRF2 signaling pathway. Free Radic Biol Med 2024; 224:770-784. [PMID: 39313014 DOI: 10.1016/j.freeradbiomed.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024]
Abstract
Gaudichaudione H (GH) is a natural small molecular compound isolated from Garcinia oligantha Merr. (Clusiaceae). Being an uncommon rare caged polyprenylated xanthone, the potential pharmacological functions of GH remain to be fully elucidated currently. In this study, we primarily focused on identifying potential bioavailable targets and elucidating related therapeutic actions. Herein, the network pharmacology analysis, metabolomics analysis and genome-wide mRNA transcription assay were performed firstly to predict the major pharmacological action and potential targets of GH. To confirm the hypothesis, gene knockout model was created using CRISPR/Cas9 method. The pharmacological action of GH was evaluated in vitro and in vivo. Firstly, our results of network pharmacology analysis and omics assay indicated that GH significantly activated NRF2 signaling pathway, and the function could be associated with liver disease treatment. Then, the pharmacological action of GH was evaluated in vitro and in vivo. The treatment with GH significantly increased the protein levels of NRF2 and promoted the transcription of NRF2 downstream genes. Further analysis suggested that GH regulated NRF2 through an autophagy-mediated non-canonical mechanism. Additionally, the administration of GH effectively protected the liver from carbon tetrachloride (CCl4)-induced liver fibrosis and inflammation, which depended on the activation of NRF2 in hepatic stellate cells and inflammatory cells respectively. Collectively, our findings underscore the potential therapeutic effect of GH on alleviating hepatic fibrosis and inflammation through the augmentation of NRF2 signaling pathway, providing a promising avenue for the treatment of liver fibrosis and inflammation in clinical settings.
Collapse
Affiliation(s)
- Mengjiao Shi
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Guo
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyan Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rongrong Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yetong Feng
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yaping Zhao
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chongyu Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ke Du
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Miaomiao Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian Zhang
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zongfang Li
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Dongmei Ren
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Environment and Genes Related To Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China.
| |
Collapse
|
5
|
Chaponda MM, Lam HYP. Schistosoma antigens: A future clinical magic bullet for autoimmune diseases? Parasite 2024; 31:68. [PMID: 39481080 PMCID: PMC11527426 DOI: 10.1051/parasite/2024067] [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: 07/05/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Autoimmune diseases are characterized by dysregulated immunity against self-antigens. Current treatment of autoimmune diseases largely relies on suppressing host immunity to prevent excessive inflammation. Other immunotherapy options, such as cytokine or cell-targeted therapies, have also been used. However, most patients do not benefit from these therapies as recurrence of the disease usually occurs. Therefore, more effort is needed to find alternative immune therapeutics. Schistosoma infection has been a significant public health problem in most developing countries. Schistosoma parasites produce eggs that continuously secrete soluble egg antigen (SEA), which is a known modulator of host immune responses by enhancing Th2 immunity and alleviating outcomes of Th1 and Th17 responses. Recently, SEA has shown promise in treating autoimmune disorders due to their substantial immune-regulatory effects. Despite this interest, how these antigens modulate human immunity demonstrates only limited pieces of evidence, and whether there is potential for Schistosoma antigens in other diseases in the future remains an unsolved question. This review discusses how SEA modulates human immune responses and its potential for development as a novel immunotherapeutic for autoimmune diseases. We also discuss the immune modulatory effects of other non-SEA schistosome antigens at different stages of the parasite's life cycle.
Collapse
Affiliation(s)
- Mphatso Mayuni Chaponda
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University Hualien Taiwan
| | - Ho Yin Pekkle Lam
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University Hualien Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University Hualien Taiwan
- Institute of Medical Science, Tzu Chi University Hualien Taiwan
| |
Collapse
|
6
|
Sun T, Li S, Li X, Lei Y, Wang B, Liu X, Yu S, Li N. Apigenin intervenes in liver fibrosis by regulating PKM2-HIF-1α mediated oxidative stress. Biochem Biophys Res Commun 2024; 721:150130. [PMID: 38761750 DOI: 10.1016/j.bbrc.2024.150130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
Apigenin (API) is a natural flavonoid compound with antioxidant, anti fibrotic, anti-inflammatory and other effects, but there is limited research on the effect of API on liver fibrosis. This study aims to explore the effect and potential mechanism of API on liver fibrosis induced by CCl4 in mice. The results indicate that API reduces oxidative stress levels, inhibits hepatic stellate cell (HSC) activation, and exerts anti liver fibrosis effects by regulating the PKM2-HIF-1α pathway. We observed that API alleviated liver tissue pathological damage and collagen deposition in CCl4 induced mouse liver fibrosis model, promoting the recovery of liver function in mice with liver fibrosis. In addition, the API inhibits the transition of Pyruvate kinase isozyme type M2 (PKM2) from dimer to tetramer formation by regulating the EGFR-MEK1/2-ERK1/2 pathway, thereby preventing dimer from entering the nucleus and blocking PKM2-HIF-1α access. This change leads to a decrease in malondialdehyde (MDA) and Catalase (CAT) levels and an increase in glutathione (GSH), superoxide dismutase (SOD), glutathione peroxidase (GSH-PX) levels, as well as total antioxidant capacity (T-AOC) in the liver of liver fibrosis mice. At the same time, API downregulated the expression of α-smooth muscle actin (α-SMA), Vimentin and Desmin in the liver tissue of mice with liver fibrosis, inhibited the activation of HSC, and reduced collagen deposition. These results indicate that API can inhibit HSC activation and alleviate CCl4 induced liver fibrosis by inhibiting the PKM2-HIF-1α pathway and reducing oxidative stress, laying an important foundation for the development and clinical application of API as a novel drug for treating liver fibrosis.
Collapse
Affiliation(s)
- Tao Sun
- Department of Internal Medicine, Henan Medical College, Zhengzhou, China
| | - Saifei Li
- Henan University of Chinese Medicine, School of Pharmacy, Zhengzhou, China
| | - Xiaoying Li
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Yanfei Lei
- Department of Internal Medicine, Henan Medical College, Zhengzhou, China
| | - Baoying Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xianghua Liu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Shanfa Yu
- School of Public Health, Henan Medical College, Zhengzhou, China
| | - Ningning Li
- Department of Pathology, Henan Medical College, Zhengzhou, China.
| |
Collapse
|
7
|
Liu L, Wang B, Ma Y, Sun K, Wang P, Li M, Dong J, Qin M, Li M, Wei C, Tan Y, He J, Guo K, Yu XA. A review of Phyllanthus urinaria L. in the treatment of liver disease: viral hepatitis, liver fibrosis/cirrhosis and hepatocellular carcinoma. Front Pharmacol 2024; 15:1443667. [PMID: 39185304 PMCID: PMC11341462 DOI: 10.3389/fphar.2024.1443667] [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: 06/04/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Due to the pathological production of liver disease in utility particularly complexity, the morbidity and mortality of liver disease including viral hepatitis, liver fibrosis/cirrhosis and hepatocellular carcinoma (HCC) are rapidly increasing worldwide. Considering its insidious onset, rapid progression and drug resistance, finding an effective therapy is particularly worthwhile. Phyllanthus urinaria L. (P. urinaria), an ethnic medicine, can be applied at the stages of viral hepatitis, liver fibrosis/cirrhosis and HCC, which demonstrates great potential in the treatment of liver disease. Currently, there are numerous reports on the application of P. urinaria in treating liver diseases, but a detailed analysis of its metabolites and a complete summary of its pharmacological mechanism are still scarce. In this review, the phytochemical metabolites and ethnopharmacological applications of P. urinaria are summarized. Briefly, P. urinaria mainly contains flavonoids, lignans, tannins, phenolic acids, terpenoids and other metabolites. The mechanisms of P. urinaria are mainly reflected in reducing surface antigen secretion and interfering with DNA polymerase synthesis for anti-viral hepatitis activity, reducing hepatic stellate cells activity, inflammation and oxidative stress for anti-liver fibrosis/cirrhosis activity, as well as preventing tumor proliferation, invasion and angiogenesis for anti-HCC activity via relevant signaling pathways. Accordingly, this review provides insights into the future application of natural products in the trilogy of liver diseases and will provide a scientific basis for further research and rational utilization of P. urinaria.
Collapse
Affiliation(s)
- Linhua Liu
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
- State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen lnternational Graduate School, Tsinghua University, Shenzhen, China
| | - Bing Wang
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| | - Yibo Ma
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| | - Kunhui Sun
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| | - Ping Wang
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| | - Meifang Li
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| | - Junlin Dong
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| | - Meirong Qin
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| | - Mingshun Li
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Chunshan Wei
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ying Tan
- State Key Laboratory of Chemical Oncogenomics, Institute of Biopharmaceutical and Health Engineering, Shenzhen lnternational Graduate School, Tsinghua University, Shenzhen, China
| | - Jinsong He
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Keying Guo
- Department of Biotechnology and Food Engineering, Guangdong-Technion Israel Institute of Technology, Shantou, China
| | - Xie-an Yu
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, China
| |
Collapse
|
8
|
Huang X, Wang X, Wang Y, Shen S, Chen W, Liu T, Wang P, Fan X, Liu L, Jia J, Cong M. TIMP-1 Promotes Expression of MCP-1 and Macrophage Migration by Inducing Fli-1 in Experimental Liver Fibrosis. J Clin Transl Hepatol 2024; 12:634-645. [PMID: 38993513 PMCID: PMC11233975 DOI: 10.14218/jcth.2023.00514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 07/13/2024] Open
Abstract
Background and Aims Tissue inhibitor of metalloproteinase-1 (TIMP-1) plays a role in the excessive generation of extracellular matrix in liver fibrosis. This study aimed to explore the pathways through which TIMP-1 controls monocyte chemoattractant protein-1 (MCP-1) expression and promotes hepatic macrophage recruitment. Methods Liver fibrosis was triggered through carbon tetrachloride, and an adeno-associated virus containing small interfering RNA targeting TIMP-1 (siRNA-TIMP-1) was administered to both rats and mice. We assessed the extent of fibrosis and macrophage recruitment. The molecular mechanisms regulating macrophage recruitment by TIMP-1 were investigated through transwell migration assays, luciferase reporter assays, the use of pharmacological modulators, and an analysis of extracellular vesicles (EVs). Results siRNA-TIMP-1 alleviated carbon tetrachloride-induced liver fibrosis, reducing macrophage migration and MCP-1 expression. Co-culturing macrophages with hepatic stellate cells (HSCs) post-TIMP-1 downregulation inhibited macrophage migration. In siRNA-TIMP-1-treated HSCs, microRNA-145 (miRNA-145) expression increased, while the expression of Friend leukemia virus integration-1 (Fli-1) and MCP-1 was inhibited. Downregulation of Fli-1 led to decreased MCP-1 expression, whereas Fli-1 overexpression increased MCP-1 expression within HSCs. Transfection with miRNA-145 mimics reduced the expression of both Fli-1 and MCP-1, while miRNA-145 inhibitors elevated the expression of both Fli-1 and MCP-1 in HSCs. miRNA-145 bound directly to the 3'-UTR of Fli-1, and miRNA-145-enriched EVs secreted by HSCs after TIMP-1 downregulation influenced macrophage recruitment. Conclusions TIMP-1 induces Fli-1 expression through miRNA-145, subsequently increasing MCP-1 expression and macrophage recruitment. MiRNA-145-enriched EVs from HSCs can transmit biological information and magnify the function of TIMP-1.
Collapse
Affiliation(s)
- Xiaoli Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xiaofan Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Yanhong Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
- Dongying People's Hospital, Dongying, Shandong, China
| | - Shuangjun Shen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xu Fan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| |
Collapse
|
9
|
Wang X, Liu H, Wang Y, Wang P, Yi Y, Lin Y, Li X. Novel protein C6ORF120 promotes liver fibrosis by activating hepatic stellate cells through the PI3K/Akt/mTOR pathway. J Gastroenterol Hepatol 2024; 39:1422-1430. [PMID: 38523410 DOI: 10.1111/jgh.16538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/14/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND AND AIM The role of C6ORF120 in promoting CCL4-induced hepatic fibrosis and its possible mechanisms were explored in C6orf120 knockout rats (C6orf120-/-) and LX-2 cells (a type of human hepatic stellate cell line). METHODS In vivo experiments, wild-type and C6orf120-/- rats were used to investigate the function of C6ORF120. In the in vitro experiments, C6ORF120 recombinant protein (rC6ORF120) at a concentration of 200 ng/mL was used to stimulate LX-2 cells. Sirius Red staining, Masson staining, western blotting, polymerase chain reaction, immunohistochemistry, and immunofluorescence were used to explore fibrosis-associated factors. RESULTS C6orf120-/- rats showed mild fibrosis and liver injury in the CCL4-induced liver fibrosis model. Furthermore, RNA-seq revealed that C6orf120-/- rats had less extracellular matrix deposition and activated stellate cells. Consistent with the in vivo, the rC6ORF120 induced LX-2 cell activation. Moreover, mechanistic studies revealed that the p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR levels were significantly elevated and LY294002 (a PI3K/Akt/mTOR typical pathway inhibitor) reversed the function of C6ORF120 in activating LX-2 cells. CONCLUSION C6ORF120 could activate hepatic stellate cells and promote hepatic fibrosis via the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xin Wang
- Department of Center of Integrated Traditional Chinese and Western Medicine, Peking University Ditan Teaching Hospital, Beijing, China
| | - Hui Liu
- Department of Center of Infectious Disease, Beijing Ditan Hospital; Capital Medical University, Beijing, China
| | - Yuqi Wang
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Peng Wang
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yunyun Yi
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yingying Lin
- Department of Center of Integrated Traditional Chinese and Western Medicine, Peking University Ditan Teaching Hospital, Beijing, China
| | - Xin Li
- Department of Center of Integrated Traditional Chinese and Western Medicine, Peking University Ditan Teaching Hospital, Beijing, China
- Department of Center of Integrated Traditional Chinese and Western Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Yuan S, Liu J, Yang L, Zhang X, Zhuang K, He S. Knockdown of circ_0044226 promotes endoplasmic reticulum stress-mediated autophagy and apoptosis in hepatic stellate cells via miR-4677-3p/SEC61G axis. J Bioenerg Biomembr 2024; 56:261-271. [PMID: 38421527 DOI: 10.1007/s10863-024-10007-0] [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: 11/22/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Downregulation of circ_0044226 has been demonstrated to reduce pulmonary fibrosis, but the role of circ_0044226 in liver fibrosis remains to be explored. In this work, we found that circ_0044226 expression was upregulated during liver fibrosis. Knockdown of circ_0044226 inhibited proliferation, promoted autophagy and apoptosis of hepatic stellate cell LX-2. Bioinformatic analysis and dual luciferase reporter assays confirmed the interaction between circ_0044226, miR-4677-3p and SEC61G. Mechanistically, knockdown of circ_0044226 suppressed SEC61G expression by releasing miR-4677-3p, thereby enhancing endoplasmic reticulum stress. Overexpression of SEC61G or endoplasmic reticulum stress inhibitor 4-phenylbutiric acid partially reversed the effect of knockdown circ_0044226 on LX-2 cell function. In vivo experiments showed that inhibition of circ_0044226 attenuated CCL4-induced liver fibrosis in mice. These imply that circ_0044226 may be a potential target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Shanshan Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710004, Shaanxi, China
| | - Jiaming Liu
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710004, Shaanxi, China
| | - Li Yang
- Department of Ultrasonography, Xi'an Children's Hospital, The Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Xin Zhang
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710004, Shaanxi, China
| | - Kun Zhuang
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710004, Shaanxi, China
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China.
- Clinical Medical Research Center for Digestive Diseases of Shaanxi Province (Oncology), Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
11
|
Ning Y, Dou X, Wang Z, Shi K, Wang Z, Ding C, Sang X, Zhong X, Shao M, Han X, Cao G. SIRT3: A potential therapeutic target for liver fibrosis. Pharmacol Ther 2024; 257:108639. [PMID: 38561088 DOI: 10.1016/j.pharmthera.2024.108639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Sirtuin3 (SIRT3) is a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase located in the mitochondria, which mainly regulates the acetylation of mitochondrial proteins. In addition, SIRT3 is involved in critical biological processes, including oxidative stress, inflammation, DNA damage, and apoptosis, all of which are closely related to the progression of liver disease. Liver fibrosis characterized by the deposition of extracellular matrix is a result of long termed or repeated liver damage, frequently accompanied by damaged hepatocytes, the recruitment of inflammatory cells, and the activation of hepatic stellate cells. Based on the functions and pharmacology of SIRT3, we will review its roles in liver fibrosis from three aspects: First, the main functions and pharmacological effects of SIRT3 were investigated based on its structure. Second, the roles of SIRT3 in major cells in the liver were summarized to reveal its mechanism in developing liver fibrosis. Last, drugs that regulate SIRT3 to prevent and treat liver fibrosis were discussed. In conclusion, exploring the pharmacological effects of SIRT3, especially in the liver, may be a potential strategy for treating liver fibrosis.
Collapse
Affiliation(s)
- Yan Ning
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Dou
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhichao Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kao Shi
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeping Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Ding
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang Zhong
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiyu Shao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China; The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Zhang S, Ren X, Zhang B, Lan T, Liu B. A Systematic Review of Statins for the Treatment of Nonalcoholic Steatohepatitis: Safety, Efficacy, and Mechanism of Action. Molecules 2024; 29:1859. [PMID: 38675679 PMCID: PMC11052408 DOI: 10.3390/molecules29081859] [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: 03/06/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the liver component of a cluster of conditions, while its subtype, nonalcoholic steatohepatitis (NASH), emerges as a potentially progressive liver disorder that harbors the risk of evolving into cirrhosis and culminating in hepatocellular carcinoma (HCC). NASH and cardiovascular disease (CVD) have common risk factors, but compared to liver-related causes, the most common cause of death in NASH patients is CVD. Within the pharmacological armamentarium, statins, celebrated for their lipid-modulating prowess, have now garnered attention for their expansive therapeutic potential in NASH. Evidence from a plethora of studies suggests that statins not only manifest anti-inflammatory and antifibrotic properties but also impart a multifaceted beneficial impact on hepatic health. In this review, we used "statin", "NAFLD", "NASH", and "CVD" as the major keywords and conducted a literature search using the PubMed and Web of Science databases to determine the safety and efficacy of statins in patients and animals with NASH and NAFLD, and the mechanism of statin therapy for NASH. Simultaneously, we reviewed the important role of the intestinal microbiota in statin therapy for NASH, as it is hoped that statins will provide new insights into modulating the harmful inflammatory microbiota in the gut and reducing systemic inflammation in NASH patients.
Collapse
Affiliation(s)
- Shiqin Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| | - Xiaoling Ren
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| | - Bingzheng Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| | - Tian Lan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Bing Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| |
Collapse
|
13
|
Zhang LF, Deng WQ, Huang QW, Zhang JJ, Wang Y, Zhou TJ, Xing L, Jiang HL. Vicious Cycle-Breaking Lipid Nanoparticles Remodeling Multicellular Crosstalk to Reverse Liver Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311474. [PMID: 38194906 DOI: 10.1002/adma.202311474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/22/2023] [Indexed: 01/11/2024]
Abstract
During liver fibrogenesis, the reciprocal crosstalk among capillarized liver sinusoidal endothelial cells (LSECs), activated hepatic stellate cells (HSCs), and dysfunctional hepatocytes constructs a self-amplifying vicious cycle, greatly exacerbating the disease condition and weakening therapeutic effect. Limited by the malignant cellular interactions, the previous single-cell centric treatment approaches show unsatisfactory efficacy and fail to meet clinical demand. Herein, a vicious cycle-breaking strategy is proposed to target and repair pathological cells separately to terminate the malignant progression of liver fibrosis. Chondroitin sulfate-modified and vismodegib-loaded nanoparticles (CS-NPs/VDG) are designed to efficiently normalize the fenestrae phenotype of LSECs and restore HSCs to quiescent state by inhibiting Hedgehog signaling pathway. In addition, glycyrrhetinic acid-modified and silybin-loaded nanoparticles (GA-NPs/SIB) are prepared to restore hepatocytes function by relieving oxidative stress. The results show successful interruption of vicious cycle as well as distinct fibrosis resolution in two animal models through multiregulation of the pathological cells. This work not only highlights the significance of modulating cellular crosstalk but also provides a promising avenue for developing antifibrotic regimens.
Collapse
Affiliation(s)
- Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Wen-Qi Deng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Qing-Wen Huang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiao-Jiao Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- College of Pharmacy, Yanbian University, Yanji, 133002, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
14
|
Zaid A, Ariel A. Harnessing anti-inflammatory pathways and macrophage nano delivery to treat inflammatory and fibrotic disorders. Adv Drug Deliv Rev 2024; 207:115204. [PMID: 38342241 DOI: 10.1016/j.addr.2024.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional "polarization" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.
Collapse
Affiliation(s)
- Ahmad Zaid
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel
| | - Amiram Ariel
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel.
| |
Collapse
|
15
|
Rajak S. Dynamics of cellular plasticity in non-alcoholic steatohepatitis (NASH). Biochim Biophys Acta Mol Basis Dis 2024; 1870:167102. [PMID: 38422712 DOI: 10.1016/j.bbadis.2024.167102] [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: 01/15/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
Non-alcoholic steatohepatitis (NASH) is a pathogenic stage of the broader non-alcoholic fatty liver disease (NAFLD). Histological presentation of NASH includes hepatocyte ballooning, macrophage polarization, ductular reaction, and hepatic stellate cell (HSCs) activation. At a cellular level, a heterogenous population of cells such as hepatocytes, macrophages, cholangiocytes, and HSCs undergo dramatic intra-cellular changes in response to extracellular triggers, which are termed "cellular plasticity. This dynamic switch in the cellular structure and function of hepatic parenchymal and non-parenchymal cells and their crosstalk culminates in the perpetuation of inflammation and fibrosis in NASH. This review presents an overview of our current understanding of cellular plasticity in NASH and its molecular mechanisms, along with possible targeting to develop cell-specific NASH therapies.
Collapse
Affiliation(s)
- Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India.
| |
Collapse
|
16
|
Zhang Y, Wu D, Tian X, Chen B. From hepatitis B virus infection to acute-on-chronic liver failure: The dynamic role of hepatic macrophages. Scand J Immunol 2024; 99:e13349. [PMID: 38441398 DOI: 10.1111/sji.13349] [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: 08/26/2023] [Revised: 11/15/2023] [Accepted: 12/11/2023] [Indexed: 03/07/2024]
Abstract
Acute-on-chronic liver failure (ACLF) is a progressive disease that is associated with rapid worsening of clinical symptoms and high mortality. A multicentre prospective study from China demonstrated that patients with hepatitis B virus-related ACLF (HBV-ACLF) exhibited worse clinical characteristics and higher mortality rates compared to non-HBV-ACLF patients. Immune dysregulation is closely linked to the potential mechanisms of initiation and progression of ACLF. Innate immune response, which is represented by monocytes/macrophages, is up-regulated across ACLF development. This suggests that monocytes/macrophages play an essential role in maintaining the immune homeostasis of ACLF. Information that has been published in recent years shows that the immune status and function of monocytes/macrophages vary in ACLF precipitated by different chronic liver diseases. Monocytes/macrophages have an immune activation effect in hepatitis B-precipitated-ACLF, but they exhibit an immune suppression in cirrhosis-precipitated-ACLF. Therefore, this review aims to explain whether this difference affects the clinical outcome in HBV-ACLF patients as well as the mechanisms involved. We summarize the novel findings that highlight the dynamic polarization phenotype and functional status of hepatic macrophages from the stage of HBV infection to ACLF development. Moreover, we discuss how different HBV-related liver disease tissue microenvironments affect the phenotype and function of hepatic macrophages. In summary, increasing developments in understanding the differences in immune phenotype and functional status of hepatic macrophages in ACLF patients will provide new perspectives towards the effective restoration of ACLF immune homeostasis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Dongsheng Wu
- Department of Anorectal Surgical, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaoling Tian
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Bin Chen
- Department of Hepatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
17
|
Hong W, Xiao T, Lin G, Liu C, Li H, Li Y, Hu H, Wu S, Wang S, Liang Z, Lin T, Liu J, Chen X. Structure-based design and synthesis of anti-fibrotic compounds derived from para-positioned 3,4,5-trisubstituted benzene. Bioorg Chem 2024; 144:107113. [PMID: 38232685 DOI: 10.1016/j.bioorg.2024.107113] [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: 11/17/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
Liver fibrosis is an abnormal wound-healing response to liver injuries. It can lead to liver cirrhosis, and even liver cancer and liver failure. There is a lack of treatment for liver fibrosis and it is of great importance to develop anti-fibrotic drugs. A pivotal event in the process of developing liver fibrosis is the activation of hepatic stellate cells (HSCs), in which the nuclear receptor Nur77 plays a crucial role. This study aimed to develop novel anti-fibrotic agents with Nur77 as the drug target by modifying the structure of THPN, a Nur77-binding and anti-melanoma compound. Specifically, a series of para-positioned 3,4,5-trisubstituted benzene ring compounds with long-chain backbone were generated and tested for anti-fibrotic activity. Among these compounds, compound A8 was with the most potent and Nur77-dependent inhibitory activity against TGF-β1-induced activation of HSCs. In a crystal structure analysis, compound A8 bound Nur77 in a peg-in-hole mode as THPN did but adopted a different conformation that could interfere the Nur77 interaction with AKT, which was previous shown to be important for an anti-fibrotic activity. In a cell-based assay, compound A8 indeed impeded the interaction between Nur77 and AKT leading to the stabilization of Nur77 without the activation of AKT. In a mouse model, compound A8 effectively suppressed the activation of AKT signaling pathway and up-regulated the cellular level of Nur77 to attenuate the HSCs activation and ameliorate liver fibrosis with no significant toxic side effects. Collectively, this work demonstrated that Nur77-targeting compound A8 is a promising anti-fibrotic drug candidate.
Collapse
Affiliation(s)
- Wenbin Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen, China; Cancer Research Center of Xiamen University, Xiamen, China; Xiamen Key Laboratory of Clinical Efficacy and Evidence Studies of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China; The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Tianyichen Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen, China; Cancer Research Center of Xiamen University, Xiamen, China
| | - Gang Lin
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Changqin Liu
- Xiamen Key Laboratory of Clinical Efficacy and Evidence Studies of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China; The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hailong Li
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China
| | - Yunlong Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen, China; Cancer Research Center of Xiamen University, Xiamen, China
| | - Hongyu Hu
- Xingzhi College, Zhejiang Normal University, Lanxi 321004, China
| | - Siqi Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen, China; Cancer Research Center of Xiamen University, Xiamen, China
| | - Songqing Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen, China; Cancer Research Center of Xiamen University, Xiamen, China
| | - Zhijian Liang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen, China; Cancer Research Center of Xiamen University, Xiamen, China
| | - Tianwei Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen, China; Cancer Research Center of Xiamen University, Xiamen, China.
| | - Jie Liu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| | - Xueqin Chen
- Xiamen Key Laboratory of Clinical Efficacy and Evidence Studies of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China; The First Affiliated Hospital of Xiamen University, Xiamen, China.
| |
Collapse
|
18
|
Sun YD, Zhang H, Li YM, Han JJ. Abnormal metabolism in hepatic stellate cells: Pandora's box of MAFLD related hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189086. [PMID: 38342420 DOI: 10.1016/j.bbcan.2024.189086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/25/2023] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
Metabolic associated fatty liver disease (MAFLD) is a significant risk factor for the development of hepatocellular carcinoma (HCC). Hepatic stellate cells (HSCs), as key mediators in liver injury response, are believed to play a crucial role in the repair process of liver injury. However, in MAFLD patients, the normal metabolic and immunoregulatory mechanisms of HSCs become disrupted, leading to disturbances in the local microenvironment. Abnormally activated HSCs are heavily involved in the initiation and progression of HCC. The metabolic disorders and abnormal activation of HSCs not only initiate liver fibrosis but also contribute to carcinogenesis. In this review, we provide an overview of recent research progress on the relationship between the abnormal metabolism of HSCs and the local immune system in the liver, elucidating the mechanisms of immune imbalance caused by abnormally activated HSCs in MAFLD patients. Based on this understanding, we discuss the potential and challenges of metabolic-based and immunology-based mechanisms in the treatment of MAFLD-related HCC, with a specific focus on the role of HSCs in HCC progression and their potential as targets for anti-cancer therapy. This review aims to enhance researchers' understanding of the importance of HSCs in maintaining normal liver function and highlights the significance of HSCs in the progression of MAFLD-related HCC.
Collapse
Affiliation(s)
- Yuan-Dong Sun
- Department of Interventional Radiology, Shandong Cancer Hospital and Institute Affiliated Shandong First Medical University, Shandong Academy of Medical Sciences, China
| | - Hao Zhang
- Department of Interventional Radiology, Shandong Cancer Hospital and Institute Affiliated Shandong First Medical University, Shandong Academy of Medical Sciences, China
| | - Yuan-Min Li
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, China
| | - Jian-Jun Han
- Department of Interventional Radiology, Shandong Cancer Hospital and Institute Affiliated Shandong First Medical University, Shandong Academy of Medical Sciences, China.
| |
Collapse
|
19
|
Tutusaus A, Morales A, García de Frutos P, Marí M. GAS6/TAM Axis as Therapeutic Target in Liver Diseases. Semin Liver Dis 2024; 44:99-114. [PMID: 38395061 PMCID: PMC11027478 DOI: 10.1055/a-2275-0408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
TAM (TYRO3, AXL, and MERTK) protein tyrosine kinase membrane receptors and their vitamin K-dependent ligands GAS6 and protein S (PROS) are well-known players in tumor biology and autoimmune diseases. In contrast, TAM regulation of fibrogenesis and the inflammation mechanisms underlying metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and, ultimately, liver cancer has recently been revealed. GAS6 and PROS binding to phosphatidylserine exposed in outer membranes of apoptotic cells links TAMs, particularly MERTK, with hepatocellular damage. In addition, AXL and MERTK regulate the development of liver fibrosis and inflammation in chronic liver diseases. Acute hepatic injury is also mediated by the TAM system, as recent data regarding acetaminophen toxicity and acute-on-chronic liver failure have uncovered. Soluble TAM-related proteins, mainly released from activated macrophages and hepatic stellate cells after hepatic deterioration, are proposed as early serum markers for disease progression. In conclusion, the TAM system is becoming an interesting pharmacological target in liver pathology and a focus of future biomedical research in this field.
Collapse
Affiliation(s)
- Anna Tutusaus
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Albert Morales
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), Barcelona, Comunidad de Madrid, Spain
| | - Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| |
Collapse
|
20
|
Kabiri-Arani S, Motallebi M, Taheri MA, Kheiripour N, Ardjmand A, Aghadavod E, Shahaboddin ME. The Effect of Heat-Killed Lactobacillus plantarum on Oxidative Stress and Liver Damage in Rats with Bile Duct Ligation-Induced Hepatic Fibrosis. Probiotics Antimicrob Proteins 2024; 16:196-211. [PMID: 36522610 DOI: 10.1007/s12602-022-10033-7] [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] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
This study is aimed at evaluating the effects of heat-killed Lactobacillus plantarum (L. plantarum) on cholestatic liver injury induced by bile duct ligation (BDL) in rats. Rats in the first group were healthy (normal control) and in the second group underwent abdominal incision (sham control). Rats in the third and fourth groups underwent common bile duct ligation and were treated with either oral distilled water (BDL control group) or heat-killed L. plantarum (BDL + L. plantarum) for 28 days. Finally, rats were sacrificed, blood samples were analyzed through biochemical methods, liver and ileum tissue tissues were histologically assessed, and the expression of the αSMA, TNF-α, IL-6, and IL-10 genes in the liver and ZO-1 gene in ileum tissues were assessed through real-time PCR. The levels of bilirubin, liver function enzymes, NO, MDA, and carbonyl protein in the BDL + L. plantarum group were significantly lower than in the BDL control group (P ≤ 0.05). SOD and CAT activity in BDL + L. plantarum group was significantly greater than the BDL control group 1.4 and 3.0 times, respectively (P ≤ 0.001). Moreover, in the BDL + L. plantarum group, the expression of the α-SMA, TNF-α, and IL-6 genes was significantly lower (3.1, 2.9, and 2.5 times), and IL-10 and ZO-1 genes were significantly greater than the BDL control group by 2.1 and 3.6 times, respectively (P ≤ 0.05). The histological assessment also confirmed the greater effectiveness of heat-killed L. plantarum in improving the morphology and parenchymal structure of the liver. Taken together, our results suggest that heat-killed L. plantarum strains are potential therapeutic agents for hepatic fibrosis.
Collapse
Affiliation(s)
- Shima Kabiri-Arani
- Department of Clinical Biochemistry, Faculty of Medicine, Kashan University of Medical Sciences, Pezeshk Blvd, Qotbe Ravandi Blvd, Kashan, Iran
| | - Mitra Motallebi
- Department of Immunology and Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Akhavan Taheri
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Nejat Kheiripour
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Ardjmand
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Esmat Aghadavod
- Department of Clinical Biochemistry, Faculty of Medicine, Kashan University of Medical Sciences, Pezeshk Blvd, Qotbe Ravandi Blvd, Kashan, Iran
| | - Mohammad Esmaeil Shahaboddin
- Department of Clinical Biochemistry, Faculty of Medicine, Kashan University of Medical Sciences, Pezeshk Blvd, Qotbe Ravandi Blvd, Kashan, Iran.
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
21
|
Bourinet M, Anty R, Gual P, Luci C. Roles of innate lymphoid cells in metabolic and alcohol-associated liver diseases. JHEP Rep 2024; 6:100962. [PMID: 38304237 PMCID: PMC10831956 DOI: 10.1016/j.jhepr.2023.100962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/09/2023] [Accepted: 10/25/2023] [Indexed: 02/03/2024] Open
Abstract
Innate lymphoid cells (ILCs) have been identified as potent regulators of inflammation, cell death and wound healing, which are the main biological processes involved in the progression of chronic liver disease. Obesity and chronic alcohol consumption are the leading contributors to chronic liver diseases in developed countries, due to inappropriate lifestyles. In particular, inflammation is a key factor in these liver abnormalities and promotes the development of more severe lesions such as fibrosis, cirrhosis and hepatocellular carcinoma. Opposite roles of ILC subsets have been described in the development of chronic liver disease, depending on the stage and aetiology of the disease. The heterogeneous family of ILCs encompasses cytotoxic natural killer cells, the cytokine-producing type 1, 2 and 3 ILCs and lymphoid tissue inducer cells. Dysfunction of these immune cells provokes uncontrolled inflammation and tissue damage, which are the basis for tumour development. In this review, we provide an overview of the recent and putative roles of ILC subsets in obesity and alcohol-associated liver diseases, which are currently the major contributors to end-stage liver complications such as fibrosis/cirrhosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Manon Bourinet
- Université Côte d’Azur, INSERM, U1065, C3M, Nice, France
| | - Rodolphe Anty
- Université Côte d’Azur, CHU, INSERM, U1065, C3M, Nice, France
| | - Philippe Gual
- Université Côte d’Azur, INSERM, U1065, C3M, Nice, France
| | - Carmelo Luci
- Université Côte d’Azur, INSERM, U1065, C3M, Nice, France
| |
Collapse
|
22
|
Peng ML, Zhang LJ, Luo Y, Xu SY, Long XM, Ao JL, Liao SG, Zhu QF, He X, Xu GB. Phomopsterone B Alleviates Liver Fibrosis through mTOR-Mediated Autophagy and Apoptosis Pathway. Molecules 2024; 29:417. [PMID: 38257331 PMCID: PMC10820960 DOI: 10.3390/molecules29020417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Liver fibrosis is the initial pathological process of many chronic liver diseases. Targeting hepatic stellate cell (HSC) activation is an available strategy for the therapy of liver fibrosis. We aimed to explore the anti-liver fibrosis activity and potential mechanism of phomopsterone B (PB) in human HSCs. The results showed that PB effectively attenuated the proliferation of TGF-β1-stimulated LX-2 cells in a concentration-dependent manner at doses of 1, 2, and 4 μM. Quantitative real-time PCR and Western blot assays displayed that PB significantly reduced the expression levels of α-SMA and collagen I/III. AO/EB and Hoechst33342 staining and flow cytometry assays exhibited that PB promoted the cells' apoptosis. Meanwhile, PB diminished the number of autophagic vesicles and vacuolated structures, and the LC3B fluorescent spots indicated that PB could effectively inhibit the accretion of autophagosomes in LX-2 cells. Moreover, rapamycin and MHY1485 were utilized to further investigate the effect of mTOR in autophagy and apoptosis. The results demonstrated that PB regulated autophagy and apoptosis via the mTOR-dependent pathway in LX-2 cells. In summary, this is the first evidence that PB effectively alleviates liver fibrosis in TGF-β1-stimulated LX-2 cells, and PB may be a promising candidate for the prevention of liver fibrosis.
Collapse
Affiliation(s)
- Mei-Lin Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province, Guian New District, Guiyang 550025, China
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education, Guiyang 550004, China
| | - Li-Jie Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
| | - Yan Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
| | - Shi-Ying Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
| | - Xing-Mei Long
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
| | - Jun-Li Ao
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education, Guiyang 550004, China
| | - Shang-Gao Liao
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province, Guian New District, Guiyang 550025, China
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education, Guiyang 550004, China
| | - Qin-Feng Zhu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province, Guian New District, Guiyang 550025, China
| | - Xun He
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province, Guian New District, Guiyang 550025, China
| | - Guo-Bo Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New District, Guiyang 550004, China; (M.-L.P.); (L.-J.Z.); (Y.L.); (S.-Y.X.); (X.-M.L.); (J.-L.A.); (S.-G.L.); (Q.-F.Z.)
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province, Guian New District, Guiyang 550025, China
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education, Guiyang 550004, China
| |
Collapse
|
23
|
Su HY, Lai CS, Lee KH, Chiang YW, Chen CC, Hsu PC. Prenatal exposure to low-dose di-(2-ethylhexyl) phthalate (DEHP) induces potentially hepatic lipid accumulation and fibrotic changes in rat offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115776. [PMID: 38056127 DOI: 10.1016/j.ecoenv.2023.115776] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a plasticizer that is widely used to enhance the flexibility and durability of various products. As an endocrine disruptor, DEHP can interfere with normal hormonal functions, posing substantial health risks to organisms. Given the critical role of the liver in DEHP metabolism, we investigated potential liver damage in offspring induced by prenatal exposure to low doses of DEHP in Sprague Dawley rats. Pregnant rats were divided into three groups and administered 20 or 200 μg/kg/day of DEHP or corn oil vehicle control via oral gavage from gestation days 0-20. Male rat offspring were euthanized on postnatal day 84, and blood and liver specimens were collected for analysis. We observed fibrotic changes in the livers of the exposed groups, accompanied by the proliferation and activation of hepatic stellate cells and upregulated expression of TGF-B and collagen 1A1. Additionally, an inflammatory response, characterized by increased macrophage infiltration and elevated levels of pro-inflammatory cytokines, was evident. Third, hepatic and serum triglyceride and serum cholesterol were notably increased, along with upregulated expression of lipid metabolism-related proteins, such as sterol regulatory element-binding protein-1c, acetyl-CoA carboxylase, fatty acid synthase, and diacylglycerol O-acyltransferase 1, particularly in the low-dose group. These results suggest that prenatal exposure to DEHP can disrupt lipid metabolism, resulting in hepatic lipid accumulation in the offspring. This exposure may also induce an inflammatory response that contributes to the development of liver fibrosis. Thus, even at relatively low doses, such exposure can precipitate latent liver damage in offspring.
Collapse
Affiliation(s)
- Hung-Yuan Su
- Department of Safety, Health and Environmental Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan; School of Chinese Medicine for Post Baccalaureate, I-Shou University, Kaohsiung 824, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan
| | - Ching-Shu Lai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan
| | - Kuo-Hsin Lee
- Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan
| | - Yu-Wei Chiang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan; Department of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chia-Chi Chen
- School of Chinese Medicine for Post Baccalaureate, I-Shou University, Kaohsiung 824, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan; Department of Physical Therapy, I-Shou University, Kaohsiung 824, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan
| | - Ping-Chi Hsu
- Department of Safety, Health and Environmental Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan; Department of Public Health, Kaohsiung Medical University, Kaohsiung, 807, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
24
|
Wang J, Wu Z, Chen X, Sun Y, Ma S, Weng J, Zhang Y, Dong K, Shao J, Zheng S. Network Pharmacology, Molecular Docking Analysis and Molecular Dynamics Simulation of Scutellaria baicalensis in the Treatment of Liver Fibrosis. Curr Pharm Des 2024; 30:1326-1340. [PMID: 38616754 DOI: 10.2174/0113816128297074240327090020] [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: 12/06/2023] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Traditional Chinese medicine Scutellaria Baicalensis (SB), one of the clinical firstline heat-clearing drugs, has obvious symptomatic advantages for hepatic fibrosis with dampness-heat stasis as its syndrome. We aim to predict and validate the potential mechanism of Scutellaria baicalensis active ingredients against liver fibrosis more scientifically and effectively. METHODS The underlying mechanism of Scutellaria baicalensis in inhibiting hepatic fibrosis was studied by applying network pharmacology, molecular docking and molecular dynamics simulation. Expression levels of markers in activated Hepatic Stellate Cells (HSC) after administration of three Scutellaria baicalensis extracts were determined by Western blot and Real-time PCR, respectively, in order to verify the anti-fibrosis effect of the active ingredients Results: There are 164 common targets of drugs and diseases screened and 115 signaling pathways obtained, which were mainly associated with protein phosphorylation, senescence and negative regulation of the apoptotic process. Western blot and Real-time PCR showed that Scutellaria baicalensis extracts could reduce the expression of HSC activation markers, and Oroxylin A had the strongest inhibitory effect on it. Molecular docking results showed that Oroxylin A had high binding activity to target proteins. Molecular dynamics simulation demonstrates promising stability of the Oroxylin A-AKT1 complex over the simulated MD time of 200 ns. CONCLUSION Scutellaria baicalensis active ingredients may inhibit HSC proliferation, reduce the generation of pro-inflammatory factors and block the anti-inflammatory effect of inflammatory signal transduction by inducing HSC apoptosis and senescence, thus achieving the effect of anti-fibrosis.
Collapse
Affiliation(s)
- Junrui Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhuoqing Wu
- Nanjing Foreign Language School, Nanjing, China
| | - Xiaolei Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Sun
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuyao Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingdan Weng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Keke Dong
- PharmaBlock Sciences (Nanjing), Inc, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
25
|
Sotoudeheian M. Galectin-3 and Severity of Liver Fibrosis in Metabolic Dysfunction-Associated Fatty Liver Disease. Protein Pept Lett 2024; 31:290-304. [PMID: 38715329 DOI: 10.2174/0109298665301698240404061300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/02/2024] [Accepted: 03/21/2024] [Indexed: 08/13/2024]
Abstract
Metabolic dysfunction-associated Fatty Liver Disease (MAFLD) is a chronic liver disease characterized by the accumulation of fat in the liver and hepatic steatosis, which can progress to critical conditions, including Metabolic dysfunction-associated Steatohepatitis (MASH), liver fibrosis, hepatic cirrhosis, and hepatocellular carcinoma. Galectin-3, a member of the galectin family of proteins, has been involved in cascades that are responsible for the pathogenesis and progression of liver fibrosis in MAFLD. This review summarizes the present understanding of the role of galectin-3 in the severity of MAFLD and its associated liver fibrosis. The article assesses the underlying role of galectin-3-mediated fibrogenesis, including the triggering of hepatic stellate cells, the regulation of extracellular degradation, and the modulation of immune reactions and responses. It also highlights the assessments of the potential diagnostic and therapeutic implications of galectin-3 in liver fibrosis during MAFLD. Overall, this review provides insights into the multifaceted interaction between galectin-3 and liver fibrosis in MAFLD, which could lead to the development of novel strategies for diagnosis and treatment of this prevalent liver disease.
Collapse
|
26
|
Wu K, Liu Y, Xia J, Liu J, Wang K, Liang H, Xu F, Liu D, Nie D, Tang X, Huang A, Chen C, Tang N. Loss of SLC27A5 Activates Hepatic Stellate Cells and Promotes Liver Fibrosis via Unconjugated Cholic Acid. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304408. [PMID: 37957540 PMCID: PMC10787101 DOI: 10.1002/advs.202304408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/11/2023] [Indexed: 11/15/2023]
Abstract
Although the dysregulation of bile acid (BA) composition has been associated with fibrosis progression, its precise roles in liver fibrosis is poorly understood. This study demonstrates that solute carrier family 27 member 5 (SLC27A5), an enzyme involved in BAs metabolism, is substantially downregulated in the liver tissues of patients with cirrhosis and fibrosis mouse models. The downregulation of SLC27A5 depends on RUNX family transcription factor 2 (RUNX2), which serves as a transcriptional repressor. The findings reveal that experimental SLC27A5 knockout (Slc27a5-/- ) mice display spontaneous liver fibrosis after 24 months. The loss of SLC27A5 aggravates liver fibrosis induced by carbon tetrachloride (CCI4 ) and thioacetamide (TAA). Mechanistically, SLC27A5 deficiency results in the accumulation of unconjugated BA, particularly cholic acid (CA), in the liver. This accumulation leads to the activation of hepatic stellate cells (HSCs) by upregulated expression of early growth response protein 3 (EGR3). The re-expression of hepatic SLC27A5 by an adeno-associated virus or the reduction of CA levels in the liver using A4250, an apical sodium-dependent bile acid transporter (ASBT) inhibitor, ameliorates liver fibrosis in Slc27a5-/- mice. In conclusion, SLC27A5 deficiency in mice drives hepatic fibrosis through CA-induced activation of HSCs, highlighting its significant implications for liver fibrosis treatment.
Collapse
Affiliation(s)
- Kang Wu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Yi Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Jie Xia
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Jiale Liu
- Institute of Life SciencesChongqing Medical UniversityChongqing400016China
| | - Kai Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Huijun Liang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Fengli Xu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Dina Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Dan Nie
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Xin Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| | - Chang Chen
- Institute of Life SciencesChongqing Medical UniversityChongqing400016China
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400010China
| |
Collapse
|
27
|
Liang W, Huang X, Shi J. Macrophages Serve as Bidirectional Regulators and Potential Therapeutic Targets for Liver Fibrosis. Cell Biochem Biophys 2023; 81:659-671. [PMID: 37695501 DOI: 10.1007/s12013-023-01173-w] [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: 12/23/2022] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
Liver fibrosis is a dynamic pathological process in which the structure and function of the liver abnormally change due to long-term complex inflammatory reactions and chronic liver injury caused by multiple internal and external factors. Previous studies believed that the activation of hepatic stellate cells is a critical part of the occurrence and development of liver fibrosis. However, an increasing number of studies have indicated that the macrophage plays an important role as a central regulator in liver fibrosis, and it directly affects the development and recovery of liver fibrosis. Studies of macrophages and liver fibrosis in the recent 10 years will be reviewed in this paper. This review will not only clarify the molecular mechanism of liver fibrosis regulated by macrophages but also provide new strategies and methods for ameliorating and treating liver fibrosis.
Collapse
Affiliation(s)
- Wei Liang
- Clinical Medical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| | - Xianing Huang
- Guangxi International Travel Healthcare Centre (Port Clinic of Nanning Customs District), Nanning, 530021, Guangxi, China
| | - Jingjing Shi
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
28
|
Zhang L, Zeng J, Wu H, Tian H, Song D, Wu W, Dong F. Knockdown of TXNDC5 alleviates CCL4-induced hepatic fibrosis in mice by enhancing endoplasmic reticulum stress. Am J Med Sci 2023; 366:449-457. [PMID: 37716602 DOI: 10.1016/j.amjms.2023.08.016] [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: 08/18/2022] [Revised: 04/28/2023] [Accepted: 08/01/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND Hepatic fibrosis is a common pathological process in many chronic liver diseases. TXNDC5 has been shown to be involved in the progression of renal and pulmonary fibrosis. However, the role of TXNDC5 in hepatic fibrosis is unknown. The purpose of this study is to explore the role and mechanism of TXNDC5 in hepatic fibrosis. METHODS We used TGF-β1 to activate LX-2 cells and reduced TXNDC5 expression by short hairpin RNA. Cell viability was assessed by CCK-8 assay. Cell apoptosis was analyzed by flow cytometry or Tunel assay. The fibrosis-related proteins and endoplasmic reticulum stress (ERs)-related proteins were measured by western blot. ELISA was performed to detect COL1AL levels and MMP2/9 activities in cell medium. A mouse model of hepatic fibrosis was constructed by intraperitoneal injection of CCL4. HE and Masson staining were performed to assess fibrosis in mouse liver tissue. RESULTS The results show that TXNDC5 was up-regulated in activated LX-2 cells and CCL4-induced hepatic fibrosis mice. Knockdown of TXNDC5 inhibited the viability of activated LX-2 cells and the production of collagen COL1A1. Knockdown of TXNDC5 promoted apoptosis of activated LX-2 cells. Mechanically, inhibition of TXNDC5 enhanced ERs, and the ERs inhibitor 4-Phenylbutyric acid (4-PBA) reversed the effect of TXNDC5 on activated LX-2 cells. More importantly, knockdown of TXNDC5 alleviated CCl4-induced hepatic fibrosis in mice. CONCLUSIONS Knockdown of TXNDC5 may reduce hepatic fibrosis by regulating ERs, and targeting TXNDC5 seems to be a candidate treatment for hepatic fibrosis.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Health Management, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Jieying Zeng
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Huaiyu Wu
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Hongtian Tian
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Di Song
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Weiqing Wu
- Department of Health Management, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Fajin Dong
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| |
Collapse
|
29
|
Lowe KO, Tanase CE, Maghami S, Fisher LE, Ghaemmaghami AM. Inflammatory Network of Liver Fibrosis and How It Can Be Targeted Therapeutically. IMMUNO 2023; 3:375-408. [DOI: 10.3390/immuno3040023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a complex, dynamic process associated with a broad spectrum of chronic liver diseases and acute liver failure, characterised by the dysregulated intrahepatic production of extracellular matrix proteins replacing functional liver cells with scar tissue. Fibrosis progresses due to an interrelated cycle of hepatocellular injury, triggering a persistent wound-healing response. The accumulation of scar tissue and chronic inflammation can eventually lead to cirrhosis and hepatocellular carcinoma. Currently, no therapies exist to directly treat or reverse liver fibrosis; hence, it remains a substantial global disease burden. A better understanding of the intricate inflammatory network that drives the initiation and maintenance of liver fibrosis to enable the rationale design of new intervention strategies is required. This review clarifies the most current understanding of the hepatic fibrosis cellular network with a focus on the role of regulatory T cells, and a possible trajectory for T cell immunotherapy in fibrosis treatment. Despite good progress in elucidating the role of the immune system in liver fibrosis, future work to better define the function of different immune cells and their mediators at different fibrotic stages is needed, which will enhance the development of new therapies.
Collapse
Affiliation(s)
- Kirstin O. Lowe
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK
| | - Leanne E. Fisher
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | |
Collapse
|
30
|
Xia S, Huang Y, Zhang Y, Zhang M, Zhao K, Han P, Tian D, Liao J, Liu J. Role of macrophage-to-myofibroblast transition in chronic liver injury and liver fibrosis. Eur J Med Res 2023; 28:502. [PMID: 37941043 PMCID: PMC10631085 DOI: 10.1186/s40001-023-01488-7] [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: 02/21/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Chronic liver injury contributes to liver fibrosis, which is characterized by the excessive deposition of extracellular matrix (ECM) components. ECM is mainly composed of myofibroblasts. Recently, macrophage-to-myofibroblasts transition (MMT), has been identified as a novel origin for myofibroblasts. However, the potential functions of MMT in chronic liver injury and liver fibrosis remain unknown. METHODS To clarify the transformation of fibrotic cells in hepatic fibrosis, liver specimens were collected from people at different stages in the progression of hepatic fibrosis and stained with immunofluorescence. Models of hepatic fibrosis such as the CCL4 model, HFD-induced NAFLD model, MCD-induced NAFLD model and ethanol-induced AFLD model were demonstrated and were stained with immunofluorescence. RESULTS Here, we uncovered macrophages underwent MMT in clinical liver fibrosis tissue samples and multiple animal models of chronic liver injury. MMT cells were found in specimens from patients with liver fibrosis on the basis of co-expression of macrophage (CD68) and myofibroblast (a-SMA) markers. Moreover, macrophages could transform into myofibroblasts in CCL4-induced liver fibrosis model, high-fat diet (HFD) and methionine-choline-deficient diet (MCD)-induced nonalcoholic fatty liver diseases (NAFLD) model, and ethanol-induced alcoholic fatty liver diseases (AFLD) model. In addition, we highlighted that MMT cells mainly had a predominant M2 phenotype in both human and experimental chronic liver injury. CONCLUSIONS Taken together, MMT acts a crucial role in chronic liver injury and liver fibrosis.
Collapse
Affiliation(s)
- Suhong Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yujie Huang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Mingyu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Kai Zhao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Jingmei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
31
|
Zhangdi H, Geng X, Li N, Xu R, Hu Y, Liu J, Zhang X, Qi J, Tian Y, Qiu J, Huang S, Cang X, Jin S. BMSCs alleviate liver cirrhosis by regulating Fstl1/Wnt/β-Catenin signaling pathway. Heliyon 2023; 9:e21010. [PMID: 37920508 PMCID: PMC10618771 DOI: 10.1016/j.heliyon.2023.e21010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/30/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
Researchers have shown that bone mesenchymal stem cells (BMSCs) can alleviate the progression of liver cirrhosis; however, it is unclear how exactly BMSCs function to cure liver disease. In this study, we used bioinformatics methods to assess differentially expressed genes (DEGs) in liver cirrhosis and found a significantly upregulated gene, Fstl1, in liver cirrhosis. In vivo and in vitro experiments showed that compared with those in the disease model group, the mRNA, and protein expression levels of Fstl1 were significantly reduced after BMSCs treatment, and the β-Catenin protein level was also significantly reduced after BMSCs treatment. Subsequently, we downregulated Fstl1 in activated hepatic stellate cells (HSCs) and found that Wnt and β-Catenin protein expression levels also decreased. Finally, we found that in BMSCs-treated activated HSCs, overexpression of Fstl1 reversed the inhibitory effect of BMSCs on the Wnt/β-Catenin signaling pathway to a certain extent. In summary, our results show that BMSCs can inhibit Wnt/β-Catenin signaling pathway activation by downregulating the protein expression level of Fstl1, thus alleviating cirrhosis. Therefore, targeted regulation of Fstl1 may provide a new therapeutic strategy for the progression of liver cirrhosis.
Collapse
Affiliation(s)
- Hanjing Zhangdi
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinyu Geng
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ning Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ruiling Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Hu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jingyang Liu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xu Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jihan Qi
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yingying Tian
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiawei Qiu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shiling Huang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xueyu Cang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
32
|
Li Y, Zhang T, Zhang J, Liu Q, Jia Q, Chen W, Tang Q, Xiong Y, Xia Y, Xu Y, Mo L, Huang Y, He J. Dually fibronectin/CD44-mediated nanoparticles targeted disrupt the Golgi apparatus and inhibit the hedgehog signaling in activated hepatic stellate cells to alleviate liver fibrosis. Biomaterials 2023; 301:122232. [PMID: 37418856 DOI: 10.1016/j.biomaterials.2023.122232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/19/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
Liver fibrosis is featured by activation of hepatic stellate cells (HSCs) and excessive accumulation of extracellular matrix (ECM). The Golgi apparatus in HSCs plays a vital role in synthesis and secretion of ECM proteins, while its targeted disruption in activated HSCs could be considered as a promising approach for liver fibrosis treatment. Here, we developed a multitask nanoparticle CREKA-CS-RA (CCR) to specifically target the Golgi apparatus of activated HSCs, based on CREKA (a specific ligand of fibronectin) and chondroitin sulfate (CS, a major ligand of CD44), in which retinoic acid (a Golgi apparatus-disturbing agent) chemically conjugated and vismodegib (a hedgehog inhibitor) encapsulated. Our results showed that CCR nanoparticles specifically targeted activated HSCs and preferentially accumulated in the Golgi apparatus. Systemic administration of CCR nanoparticles exhibited significantly accumulation in CCl4-induced fibrotic liver, which was attributed to specific recognition with fibronectin and CD44 on activated HSCs. CCR nanoparticles loaded with vismodegib not only disrupted Golgi apparatus structure and function but also inhibited the hedgehog signaling pathway, thus markedly suppressing HSC activation and ECM secretion in vitro and in vivo. Moreover, vismodegib-loaded CCR nanoparticles effectively inhibited the fibrogenic phenotype in CCl4-induced liver fibrosis mice without causing obvious toxicity. Collectively, these findings indicate that this multifunctional nanoparticle system can effectively deliver therapeutic agents to the Golgi apparatus of activated HSCs, thus has potential treatment of liver fibrosis with minimal side effects.
Collapse
Affiliation(s)
- Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ting Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jinhang Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qinhui Liu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingyi Jia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wenfei Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qin Tang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yimin Xiong
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yan Xia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan Province, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
33
|
Hassan GS, Flores Molina M, Shoukry NH. The multifaceted role of macrophages during acute liver injury. Front Immunol 2023; 14:1237042. [PMID: 37736102 PMCID: PMC10510203 DOI: 10.3389/fimmu.2023.1237042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
The liver is situated at the interface of the gut and circulation where it acts as a filter for blood-borne and gut-derived microbes and biological molecules, promoting tolerance of non-invasive antigens while driving immune responses against pathogenic ones. Liver resident immune cells such as Kupffer cells (KCs), a subset of macrophages, maintain homeostasis under physiological conditions. However, upon liver injury, these cells and others recruited from circulation participate in the response to injury and the repair of tissue damage. Such response is thus spatially and temporally regulated and implicates interconnected cells of immune and non-immune nature. This review will describe the hepatic immune environment during acute liver injury and the subsequent wound healing process. In its early stages, the wound healing immune response involves a necroinflammatory process characterized by partial depletion of resident KCs and lymphocytes and a significant infiltration of myeloid cells including monocyte-derived macrophages (MoMFs) complemented by a wave of pro-inflammatory mediators. The subsequent repair stage includes restoring KCs, initiating angiogenesis, renewing extracellular matrix and enhancing proliferation/activation of resident parenchymal and mesenchymal cells. This review will focus on the multifaceted role of hepatic macrophages, including KCs and MoMFs, and their spatial distribution and roles during acute liver injury.
Collapse
Affiliation(s)
- Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Manuel Flores Molina
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
34
|
Huang H, Hou J, Liao Y, Wei F, Xing B. Polyethylene microplastics impede the innate immune response by disrupting the extracellular matrix and signaling transduction. iScience 2023; 26:107390. [PMID: 37554443 PMCID: PMC10405319 DOI: 10.1016/j.isci.2023.107390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 06/01/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023] Open
Abstract
Microplastics (MPs) can accumulate in animal organs. Numerous studies have linked MPs with immune system. However, the impact of MPs on immune response remains unclear. This study examined the innate immune response of mice exposed to 5 μm MPs. In the lipopolysaccharide challenge, mice treated with MPs exhibited lower levels of serum immune factors and activated immune cells. MPs disrupted immune-related receptors and cause dysfunction in cell signal transduction within the liver and spleen. Proteomic analysis revealed that MPs impede the activation of serum immune-related signals. In addition, the tissue section imaging exhibited a significant enrichment of MPs in the extracellular matrix (ECM), consistent with the ECM dysfunction and immune receptor suppression. Therefore, our data suggest excessive MPs accumulation in ECM inhibits cell signaling pathways, thereby suppressing the activation of immune responses. We propose the biotoxicity of MPs is partly through the MP disruption of ECM (MPDEM).
Collapse
Affiliation(s)
- Haipeng Huang
- School of Life Science, Tsinghua University, Beijing 100084, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiaqi Hou
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Yilie Liao
- School of Life Science, Tsinghua University, Beijing 100084, China
| | - Fangchao Wei
- School of Life Science, Tsinghua University, Beijing 100084, China
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708, USA
| | - Baoshan Xing
- Stockbridge School of Agriculture, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
35
|
Wang Y, Shi K, Tu J, Ke C, Chen N, Wang B, Liu Y, Zhou Z. Atractylenolide III Ameliorates Bile Duct Ligation-Induced Liver Fibrosis by Inhibiting the PI3K/AKT Pathway and Regulating Glutamine Metabolism. Molecules 2023; 28:5504. [PMID: 37513376 PMCID: PMC10383814 DOI: 10.3390/molecules28145504] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Liver fibrosis is one of the leading causes of hepatic sclerosis and hepatocellular carcinoma worldwide. However, the complex pathophysiological mechanisms of liver fibrosis are unknown, and no specific drugs are available to treat liver fibrosis. Atractylenolide III (ATL III) is a natural compound isolated from the plant Atractylodes lancea (Thunb.) DC. that possesses antioxidant properties and the ability to inhibit inflammatory responses. In this study, cholestatic hepatic fibrosis was induced in mice using a bile duct ligation (BDL) model and treated with 10 mg/kg and 50 mg/kg of ATL III via gavage for 14 days. ATL III significantly reduced the liver index, lowered serum ALT and AST levels, and reduced liver injury in bile-duct-ligated mice. In addition, ATL III significantly attenuated histopathological changes and reduced collagen deposition. ATL III reduced the expression of fibrosis-related genes α-smooth muscle actin (α-SMA), Collagen I (col1a1), Collagen IV (col4a2), and fibrosis-related proteins α-SMA and col1a1 in liver tissue. Using RNA sequencing (RNA-seq) to screen molecular targets and pathways, ATL III was found to affect the PI3K/AKT singling pathway by inhibiting the phosphorylation of PI3K and AKT, thereby ameliorating BDL-induced liver fibrosis. Gas chromatography-mass spectrometry (GC-MS) was used to evaluate the effect of ATL III on liver metabolites in BDL mice. ATL III further affected glutamine metabolism by down-regulating the activity of glutamine (GLS1) and glutamine metabolism. ATL III further affected glutamine metabolism by down-regulating the activity of glutaminase (GLS1), as well as glutamine metabolism. Therefore, we conclude that ATL III attenuates liver fibrosis by inhibiting the PI3K/AKT pathway and glutamine metabolism, suggesting that ATL III is a potential drug candidate for treating liver fibrosis.
Collapse
Affiliation(s)
- Yan Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Kun Shi
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Jiyuan Tu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Chang Ke
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Niping Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Bo Wang
- Hubei Institute for Drug Control, NMPA Key Laboratory of Quality Control of Chinese Medicine, Hubei Engineering Research Center for Drug Quality Control, Wuhan 430075, China
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Zhongshi Zhou
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| |
Collapse
|
36
|
Zhao Y, Wang H, He T, Ma B, Chen G, Tzeng C. Knockdown of Yap attenuates TAA-induced hepatic fibrosis by interaction with hedgehog signals. J Cell Commun Signal 2023:10.1007/s12079-023-00775-6. [PMID: 37338798 DOI: 10.1007/s12079-023-00775-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
Liver fibrosis is an aberrant wound healing response to tissue injury characterized by excessive extracellular matrix deposition and loss of normal liver architecture. Hepatic stellate cells (HSCs) activation is regards to be the major process in liver fibrogenesis which is dynamic and reversible. Both Hippo signaling core factor Yap and Hedgehog (Hh) signaling promote HSCs transdifferentiation thereby regulating the repair process of liver injury. However, the molecular function of YAP and the regulation between Yap and Hh during fibrogenesis remain uncertain. In this study, the essential roles of Yap in liver fibrosis were investigated. Yap was detected to be increased in liver fibrotic tissue by the thioacetamide (TAA)-induced zebrafish embryonic and adult models. Inhibition of Yap by both embryonic morpholino interference and adult's inhibitor treatment was proved to alleviate TAA-induced liver lesions by and histology and gene expression examination. Transcriptomic analysis and gene expression detection showed that Yap and Hh signaling pathway have a cross talking upon TAA-induced liver fibrosis. In addition, TAA induction promoted the nuclear colocalization of YAP and Hh signaling factor GLI2α. This study demonstrates that Yap and Hh play synergistic protective roles in liver fibrotic response and provides new theoretical insight concerning the mechanisms of fibrosis progression.
Collapse
Affiliation(s)
- Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China.
| | - Huiling Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Tianhua He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Guoguang Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Chimeng Tzeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005, China.
- Translational Medicine Research Center-Key Laboratory for Cancer T-Cell Theragnostic and Clinical Translation, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China.
- Xiamen Chang Gung Hospital Medical Research Center, Xiamen, Fujian, China.
| |
Collapse
|
37
|
Yum YJ, Yoo J, Bang K, Jun JE, Jeong IK, Ahn KJ, Chung HY, Hwang YC. Peroxisome proliferator-activated receptor γ activation ameliorates liver fibrosis-differential action of transcription factor EB and autophagy on hepatocytes and stellate cells. Hepatol Commun 2023; 7:e0154. [PMID: 37204406 PMCID: PMC10538880 DOI: 10.1097/hc9.0000000000000154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/07/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Peroxisome proliferator-activated receptor γ (PPARγ) activation suppresses HSC activation and liver fibrosis. Moreover, autophagy is implicated in hepatic lipid metabolism. Here, we determined whether PPARγ activation ameliorates HSC activation by downregulating transcription factor EB (TFEB)-mediated autophagy. METHODS AND RESULTS Atg7 or Tfeb knockdown in human HSC line LX-2 cells downregulated the expression of fibrogenic markers including α smooth muscle actin, glial fibrillary acidic protein, and collagen type 1. Conversely, Atg7 or Tfeb overexpression upregulated fibrogenic marker expression. Rosiglitazone (RGZ)-mediated PPARγ activation and/or overexpression in LX-2 cells and primary HSCs decreased autophagy, as indicated by LC3B conversion, total and nuclear-TFEB contents, mRFP-LC3 and BODIPY 493/503 colocalization, and GFP-LC3 and LysoTracker colocalization. RGZ treatment decreased liver fat content, liver enzyme levels, and fibrogenic marker expression in high-fat high-cholesterol diet-fed mice. Electron microscopy showed that RGZ treatment restored the high-fat high-cholesterol diet-mediated lipid droplet decrease and autophagic vesicle induction in primary HSCs and liver tissues. However, TFEB overexpression in LX-2 cells offset the aforementioned effects of RGZ on autophagic flux, lipid droplets, and fibrogenic marker expression. CONCLUSIONS Activation of PPARγ with RGZ ameliorated liver fibrosis and downregulation of TFEB and autophagy in HSCs may be important for the antifibrotic effects of PPARγ activation.
Collapse
|
38
|
Wang Z, Du K, Jin N, Tang B, Zhang W. Macrophage in liver Fibrosis: Identities and mechanisms. Int Immunopharmacol 2023; 120:110357. [PMID: 37224653 DOI: 10.1016/j.intimp.2023.110357] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
Liver fibrosis is a chronic disease characterized by the deposition of extracellular matrix and continuous loss of tissues that perform liver functions. Macrophages are crucial modulators of innate immunity and play important roles in liver fibrogenesis. Macrophages comprise heterogeneous subpopulations that exhibit different cellular functions. Understanding the identity and function of these cells is essential for understanding the mechanisms of liver fibrogenesis. According to different definitions, liver macrophages are divided into M1/M2 macrophages or monocyte-derived macrophages/Kupffer cells. Classic M1/M2 phenotyping corresponds to pro- or anti-inflammatory effects, and, therefore, influences the degree of fibrosis in later phases. In contrast, the origin of the macrophages is closely associated with their replenishment and activation during liver fibrosis. These two classifications of macrophages depict the function and dynamics of liver-infiltrating macrophages. However, neither description properly elucidates the positive or negative role of macrophages in liver fibrosis. Critical tissue cells mediating liver fibrosis include hepatic stellate cells and hepatic fibroblasts, with hepatic stellate cells being of particular interest because of their close association with macrophages in liver fibrosis. However, the molecular biological descriptions of macrophages are inconsistent between mice and humans, warranting further investigations. In liver fibrosis, macrophages can secrete various pro-fibrotic cytokines, such as TGF-β, Galectin-3 and interleukins (ILs), and fibrosis-inhibiting cytokines, such as IL10. These different secretions may be associated with the specific identity and spatiotemporal characteristics of macrophages. Furthermore, during fibrosis dissipation, macrophages may degrade extracellular matrix by secreting matrix metalloproteinases (MMPs). Notably, using macrophages as therapeutic targets in liver fibrosis has been explored. The current therapeutic approaches for liver fibrosis can by categorized as follows: treatment with macrophage-related molecules and macrophage infusion therapy. Although there have been limited studies, macrophages have shown reliable potential for liver fibrosis treatment. In this review, we focu on the identity and function of macrophages and their relationship to the progression and regression of liver fibrosis.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Emergency Medicine Center, Jinhua Municipal Central Hospital, Zhejiang, China.
| | - Kailei Du
- Dongyang Peoples hospital, Zhejiang, China
| | - Nake Jin
- Ningbo Hangzhou Bay Hospital, Zhejiang, China
| | - Biao Tang
- Jinhua Municipal Central Hospital, Zhejiang, China
| | - Wenwu Zhang
- Department of Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
39
|
Zhao YQ, Deng XW, Xu GQ, Lin J, Lu HZ, Chen J. Mechanical homeostasis imbalance in hepatic stellate cells activation and hepatic fibrosis. Front Mol Biosci 2023; 10:1183808. [PMID: 37152902 PMCID: PMC10157180 DOI: 10.3389/fmolb.2023.1183808] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023] Open
Abstract
Chronic liver disease or repeated damage to hepatocytes can give rise to hepatic fibrosis. Hepatic fibrosis (HF) is a pathological process of excessive sedimentation of extracellular matrix (ECM) proteins such as collagens, glycoproteins, and proteoglycans (PGs) in the hepatic parenchyma. Changes in the composition of the ECM lead to the stiffness of the matrix that destroys its inherent mechanical homeostasis, and a mechanical homeostasis imbalance activates hepatic stellate cells (HSCs) into myofibroblasts, which can overproliferate and secrete large amounts of ECM proteins. Excessive ECM proteins are gradually deposited in the Disse gap, and matrix regeneration fails, which further leads to changes in ECM components and an increase in stiffness, forming a vicious cycle. These processes promote the occurrence and development of hepatic fibrosis. In this review, the dynamic process of ECM remodeling of HF and the activation of HSCs into mechanotransduction signaling pathways for myofibroblasts to participate in HF are discussed. These mechanotransduction signaling pathways may have potential therapeutic targets for repairing or reversing fibrosis.
Collapse
Affiliation(s)
- Yuan-Quan Zhao
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xi-Wen Deng
- Graduate School of Youjiang Medical University for Nationalities, Baise, China
| | - Guo-Qi Xu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jie Lin
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hua-Ze Lu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jie Chen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
40
|
Xiong Y, Wu B, Guo X, Shi D, Xia H, Xu H, Liu X. Galangin delivered by retinoic acid-modified nanoparticles targeted hepatic stellate cells for the treatment of hepatic fibrosis. RSC Adv 2023; 13:10987-11001. [PMID: 37033441 PMCID: PMC10077338 DOI: 10.1039/d2ra07561j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/17/2023] [Indexed: 04/11/2023] Open
Abstract
Hepatic fibrosis (HF) is a chronic hepatic pathological process induced by various liver injuries, with few available therapies. Previous research studies revealed that HF is characterized by the accumulation of excess extracellular matrix in the liver, mainly overexpressed by activated hepatic stellate cells (HSC). Therefore, HSC have been targeted in clinical trials for the management of HF. The aim of the present study was to develop an anti-HF drug delivery system with acrylic resin (Eudragit® RS100, Eud RS100) nanoparticles (NPs) through modification by retinoic acid (RA), modified for binding the retinol-binding protein reporter (RBPR) in HSC. Galangin (GA), is a multiple effects flavonoid which has demonstrated an anti-HF effect in our previous studies. In this study, GA was utilized for the treatment of HF. The results revealed that the NPs were well formed (diameter: 70 nm), spherical in shape, and exhibited uniform distribution and a high encapsulation efficiency. Moreover, a prominent controlled release effect and a significant increase in bioavailability was observed following the encapsulation of GA in NPs. These findings indicated that the limitation of low bioavailability due to the hydrophobic feature of GA was overcome. Furthermore, the pharmacodynamics studies demonstrated that NPs could drastically influence the anti-HF effects of GA after modification with retinoic acid. The results of the present study suggested that retinoic acid-modified GA NPs represent a promising candidate in the development of an anti-HF drug delivery system for the treatment of HF.
Collapse
Affiliation(s)
- Yuanguo Xiong
- Department of Pharmacy, Renmin Hospital of Wuhan University Wuhan 430060 China
- School of Pharmaceuticals, Hubei University of Chinese Medicine, No. 1 HuangJiahu Road West Wuhan 430065 China +86 27 68890239
| | - Bing Wu
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine Shiyan 442000 China
- Department of Cardiology, Renmin Hospital of Wuhan University Jiefang Road 238 Wuhan 430060 China +86 27 88041911 +86 27 88041911
- Cardiovascular Research Institute, Wuhan University Jiefang Road 238 Wuhan 430060 China
- Hubei Key Laboratory of Cardiology Wuhan 430060 China
| | - Xianxi Guo
- Department of Pharmacy, Renmin Hospital of Wuhan University Wuhan 430060 China
| | - Dong Shi
- School of Pharmaceuticals, Hubei University of Chinese Medicine, No. 1 HuangJiahu Road West Wuhan 430065 China +86 27 68890239
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University Jiefang Road 238 Wuhan 430060 China +86 27 88041911 +86 27 88041911
- Cardiovascular Research Institute, Wuhan University Jiefang Road 238 Wuhan 430060 China
- Hubei Key Laboratory of Cardiology Wuhan 430060 China
| | - Hanlin Xu
- School of Pharmaceuticals, Hubei University of Chinese Medicine, No. 1 HuangJiahu Road West Wuhan 430065 China +86 27 68890239
| | - Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University Jiefang Road 238 Wuhan 430060 China +86 27 88041911 +86 27 88041911
- Cardiovascular Research Institute, Wuhan University Jiefang Road 238 Wuhan 430060 China
- Hubei Key Laboratory of Cardiology Wuhan 430060 China
| |
Collapse
|
41
|
Li QY, Gong T, Huang YK, Kang L, Warner CA, Xie H, Chen LM, Duan XQ. Role of noncoding RNAs in liver fibrosis. World J Gastroenterol 2023; 29:1446-1459. [PMID: 36998425 PMCID: PMC10044853 DOI: 10.3748/wjg.v29.i9.1446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/27/2022] [Accepted: 02/27/2023] [Indexed: 03/07/2023] Open
Abstract
Liver fibrosis is a wound-healing response following chronic liver injury caused by hepatitis virus infection, obesity, or excessive alcohol. It is a dynamic and reversible process characterized by the activation of hepatic stellate cells and excess accumulation of extracellular matrix. Advanced fibrosis could lead to cirrhosis and even liver cancer, which has become a significant health burden worldwide. Many studies have revealed that noncoding RNAs (ncRNAs), including microRNAs, long noncoding RNAs and circular RNAs, are involved in the pathogenesis and development of liver fibrosis by regulating signaling pathways including transforming growth factor-β pathway, phosphatidylinositol 3-kinase/protein kinase B pathway, and Wnt/β-catenin pathway. NcRNAs in serum or exosomes have been reported to tentatively applied in the diagnosis and staging of liver fibrosis and combined with elastography to improve the accuracy of diagnosis. NcRNAs mimics, ncRNAs in mesenchymal stem cell-derived exosomes, and lipid nanoparticles-encapsulated ncRNAs have become promising therapeutic approaches for the treatment of liver fibrosis. In this review, we update the latest knowledge on ncRNAs in the pathogenesis and progression of liver fibrosis, and discuss the potentials and challenges to use these ncRNAs for diagnosis, staging and treatment of liver fibrosis. All these will help us to develop a comprehensive understanding of the role of ncRNAs in liver fibrosis.
Collapse
Affiliation(s)
- Qing-Yuan Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Tao Gong
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Yi-Ke Huang
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| | - Lan Kang
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| | - Charlotte A Warner
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - He Xie
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xi’an 710077, Shaanxi Province, China
| | - Li-Min Chen
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xi’an 710077, Shaanxi Province, China
| | - Xiao-Qiong Duan
- Center for Transfusion-transmitted Infectious Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, Sichuan Province, China
| |
Collapse
|
42
|
Zhao Q, Liang L, Zhai F, Ling G, Xiang R, Jiang X. A bibliometric and visualized analysis of liver fibrosis from 2002 to 2022. J Gastroenterol Hepatol 2023; 38:359-369. [PMID: 36459993 DOI: 10.1111/jgh.16081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022]
Abstract
Fibrosis of the liver is a degenerative alteration that occurs in the majority of chronic liver disorders. Further progression can lead to cirrhosis, liver failure, and hepatocellular carcinoma, which can seriously affect the health and lives of patients. The field of liver fibrosis research has flourished in the last 20 years, with approximately 9000 articles retrieved from the Web of Science Core Collection database alone. In order to identify future research hotspots and potential paths in a thorough and scientifically reliable manner, it is important to organize and visualize the research on this topic from a holistic and very general perspective. This study used bibliometric analysis with CiteSpace and VOSviewer software to provide a quantitative analysis, hotspot mining, and commentary of articles published in the field of liver fibrosis over the last 20 years. This bibliometric analysis contains a total of 8994 articles with 45667 authors from 6872 institutions in 97 countries, published in 1371 journals and citing 156 309 references. The literature volume has steadily increased over the last 20 years. Research has focused on gastroenterology and hepatology, pharmacology and pharmacy, and medicine, research, and experimental areas. We found that the pathological mechanisms, diagnostic and quantitative methods, etiology, and antifibrotic strategies constitute the knowledge structure of liver fibrosis. Finding mechanisms for liver fibrosis regression, identifying precise noninvasive diagnostic and prognostic biomarkers, and creating efficient liver fibrosis patient treatments are the main goals of current research.
Collapse
Affiliation(s)
- Qianqian Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Luhua Liang
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Fei Zhai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Guixia Ling
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Rongwu Xiang
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.,Liaoning Professional Technology Innovation Center on Medical Big Data and Artificial Intelligence, Shenyang, 110016, China
| | - Xiwei Jiang
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
43
|
Research progress of metformin in the treatment of liver fibrosis. Int Immunopharmacol 2023; 116:109738. [PMID: 36696857 DOI: 10.1016/j.intimp.2023.109738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/24/2023]
Abstract
Liver fibrosis is a disease with significant morbidity and mortality. It is a chronic pathological process characterized by an imbalance of extracellular matrix production and degradation in liver tissue. Metformin is a type of hypoglycemic biguanide drug, which can be used in the treatment of liver fibrosis, but its anti-fibrotic effect and mechanism of action are unclear. The purpose of this article is to review the research progress of metformin in the treatment of liver fibrosis and to provide a theoretical basis for its application in the treatment of liver fibrosis.
Collapse
|
44
|
Zhao J, Yang Q, Liu Z, Xu P, Tian L, Yan J, Li K, Lin B, Bian L, Xi Z, Liu X. The impact of subchronic ozone exposure on serum metabolome and the mechanisms of abnormal bile acid and arachidonic acid metabolisms in the liver. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114573. [PMID: 36701875 DOI: 10.1016/j.ecoenv.2023.114573] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 06/17/2023]
Abstract
Ambient ozone (O3) pollution can induce respiratory and cardiovascular toxicity. However, its impact on the metabolome and the underlying mechanisms remain unclear. This study first investigated the serum metabolite changes in rats exposed to 0.5 ppm O3 for 3 months using untargeted metabolomic approach. Results showed chronic ozone exposure significantly altered the serum levels of 34 metabolites with potential increased risk of digestive, respiratory and cardiovascular disease. Moreover, bile acid synthesis and secretion, and arachidonic acid (AA) metabolism became the most prominent affected metabolic pathways after O3 exposure. Further studies on the mechanisms found that the elevated serum toxic bile acid was not due to the increased biosynthesis in the liver, but the reduced reuptake from the portal vein to hepatocytes owing to repressed Ntcp and Oatp1a1, and the decreased bile acid efflux in hepatocytes as a results of inhibited Bsep, Ostalpha and Ostbeta. Meanwhile, decreased expressions of detoxification enzyme of SULT2A1 and the important regulators of FXR, PXR and HNF4α also contributed to the abnormal bile acids. In addition, O3 promoted the conversion of AA into thromboxane A2 (TXA2) and 20-hydroxyarachidonic acid (20-HETE) in the liver by up-regulation of Fads2, Cyp4a and Tbxas1 which resulting in decreased AA and linoleic acid (LA), and increased thromboxane B2 (TXB2) and 20-HETE in the serum. Furthermore, apparent hepatic chronic inflammation, fibrosis and abnormal function were found in ozone-exposed rats. These results indicated chronic ozone exposure could alter serum metabolites by interfering their metabolism in the liver, and inducing liver injury to aggravate metabolic disorders.
Collapse
Affiliation(s)
- Jiao Zhao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Qingcheng Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Zhiyuan Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Pengfei Xu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Jun Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Liping Bian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Xiaohua Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| |
Collapse
|
45
|
Carpentier N, Urbani L, Dubruel P, Van Vlierberghe S. The native liver as inspiration to create superior in vitro hepatic models. Biomater Sci 2023; 11:1091-1115. [PMID: 36594602 DOI: 10.1039/d2bm01646j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drug induced liver injury (DILI) is one of the major reasons of drug withdrawal during the different phases of drug development. The later in the drug development a drug is discovered to be toxic, the higher the economical as well as the ethical impact will be. In vitro models for early detection of drug liver toxicity are under constant development, however to date a superior model of the liver is still lacking. Ideally, a highly reliable model should be established to maintain the different hepatic cell functionalities to the greatest extent possible, during a period of time long enough to allow for tracking of the toxicity of compounds. In the case of DILI, toxicity can appear even after months of exposure. To reach this goal, an in vitro model should be developed that mimics the in vivo liver environment, function and response to external stimuli. The different approaches for the development of liver models currently used in the field of tissue engineering will be described in this review. Combining different technologies, leading to optimal materials, cells and 3D-constructs will ultimately lead to an ideal superior model that fully recapitulates the liver.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
46
|
Choi J, Cho Y, Choi H, Lee S, Han H, Lee J, Kwon J. Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways. Int J Mol Sci 2023; 24:ijms24043439. [PMID: 36834849 PMCID: PMC9959661 DOI: 10.3390/ijms24043439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 02/11/2023] Open
Abstract
Risk signals are characteristic of many common inflammatory diseases and can function to activate nucleotide-binding oligomerization (NLR) family pyrin domain-containing 3 (NLRP3), the innate immune signal receptor in cytoplasm. The NLRP3 inflammasome plays an important role in the development of liver fibrosis. Activated NLRP3 nucleates the assembly of inflammasomes, leading to the secretion of interleukin (IL)-1β and IL-18, the activation of caspase-1, and the initiation of the inflammatory process. Therefore, it is essential to inhibit the activation of the NLRP3 inflammasome, which plays a vital role in the immune response and in initiating inflammation. RAW 264.7 and LX-2 cells were primed with lipopolysaccharide (LPS) for 4 h and subsequently stimulated for 30 min with 5 mM of adenosine 5'-triphosphate (ATP) to activate the NLRP3 inflammasome. Thymosin beta 4 (Tβ4) was supplemented to RAW264.7 and LX-2 cells 30 min before ATP was added. As a result, we investigated the effects of Tβ4 on the NLRP3 inflammasome. Tβ4 prevented LPS-induced NLRP3 priming by inhibiting NF-kB and JNK/p38 MAPK expression and the LPS and ATP-induced production of reactive oxygen species. Moreover, Tβ4 induced autophagy by controlling autophagy markers (LC3A/B and p62) through the inhibition of the PI3K/AKT/mTOR pathway. LPS combined with ATP significantly increased thee protein expression of inflammatory mediators and NLRP3 inflammasome markers. These events were remarkably suppressed by Tβ4. In conclusion, Tβ4 attenuated NLRP3 inflammasomes by inhibiting NLRP3 inflammasome-related proteins (NLRP3, ASC, IL-1β, and caspase-1). Our results indicate that Tβ4 attenuated the NLRP3 inflammasome through multiple signaling pathway regulations in macrophage and hepatic stellate cells. Therefore, based on the above findings, it is hypothesized that Tβ4 could be a potential inflammatory therapeutic agent targeting the NLRP3 inflammasome in hepatic fibrosis regulation.
Collapse
|
47
|
Yu M, He X, Song X, Gao J, Pan J, Zhou T, Wang Q, Zhu W, Ma H, Zeng H, Xu C, Yu C. Biglycan promotes hepatic fibrosis through activating heat shock protein 47. Liver Int 2023; 43:500-512. [PMID: 36371672 DOI: 10.1111/liv.15477] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/07/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Biglycan (BGN) is a small leucine-rich proteoglycan that participates in the production of excess extracellular matrix (ECM) and is related to fibrosis in many organs. However, the role of BGN in liver fibrosis remains poorly understood. This study aimed to investigate the role and mechanism of BGN in liver fibrosis. METHODS Human liver samples, Bgn-/0 (BGN KO) mice and a human LX-2 hepatic stellate cells (HSCs) model were applied for the study of experimental fibrosis. GEO data and single-cell RNA-seq data of human liver tissue were analysed as a bioinformatic approach. Coimmunoprecipitation, immunofluorescence staining, western blotting and qRT-PCR were conducted to identify the regulatory effects of BGN on heat shock protein 47 (HSP47) expression and liver fibrosis. RESULTS We observed that hepatic BGN expression was significantly increased in patients with fibrosis and in a mouse model of liver fibrosis. Genetic deletion of BGN disrupted TGF-β1 pathway signalling and alleviated liver fibrosis in mice administered carbon tetrachloride (CCl4 ). siRNA-mediated knockdown of BGN significantly reduced TGF-β1-induced ECM deposition and fibroblastic activation in LX-2 cells. Mechanistically, BGN directly interacted with and positively regulated the collagen synthesis chaperon protein HSP47. Rescue experiments showed that BGN promoted hepatic fibrosis by regulating ECM deposition and HSC activation by positively regulating HSP47. CONCLUSION Our data indicate that BGN promotes hepatic fibrosis by regulating ECM deposition and HSC activation through an HSP47-dependent mechanism. BGN may be a new biomarker of hepatic fibrosis and a novel target for disease prevention and treatment.
Collapse
Affiliation(s)
- Mengli Yu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinjue He
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Song
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianguo Gao
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Pan
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianyu Zhou
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qinqiu Wang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huijian Ma
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hang Zeng
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengfu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Du TY, Gao YX, Zheng YS. Identification of key genes related to immune infiltration in cirrhosis via bioinformatics analysis. Sci Rep 2023; 13:1876. [PMID: 36725885 PMCID: PMC9892033 DOI: 10.1038/s41598-022-26794-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/20/2022] [Indexed: 02/03/2023] Open
Abstract
Cirrhosis is the most common subclass of liver disease worldwide and correlated to immune infiltration. However, the immune-related molecular mechanism underlying cirrhosis remains obscure. Two gene expression profiles GSE89377 and GSE139602 were investigated to identify differentially expressed genes (DEGs) related to cirrhosis. Enrichment analysis for DEGs was conducted. Next, the immune infiltration of DEGs was evaluated using CIBERSORT algorithm. The hub DEGs with tight connectivity were identified using the String and Cytoscape databases, and the expression difference of these hub genes between normal liver and cirrhosis samples was determined. Moreover, in order to evaluate the discriminatory ability of hub genes and obtained the area under the receiver operating characteristic curve values in the GSE89377 and GSE139602 datasets. Finally, the association between hub DEGs and immune cell infiltration was explored by Spearman method. Among the 299 DEGs attained, 136 were up-regulated and 163 were down-regulated. Then the enrichment function analysis of DEGs and CIBERSORT algorithm showed significant enrichment in immune and inflammatory responses. And four hub DEGs (ACTB, TAGLN, VIM, SOX9) were identified, which also showed a diagnostic value in the GSE89377 and GSE 139,602 datasets. Finally, the immune infiltration analysis indicated that, these hub DEGs were highly related to immune cells. This study revealed key DEGs involved in inflammatory immune responses of cirrhosis, which could be used as biomarkers for diagnosis or therapeutic targets of cirrhosis.
Collapse
Affiliation(s)
- Tong-Yue Du
- Department of Critical Care Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.-1-1, Zhongfu Road, Nanjing, 210003, China
| | - Ya-Xian Gao
- Department of Critical Care Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.-1-1, Zhongfu Road, Nanjing, 210003, China
| | - Yi-Shan Zheng
- Department of Critical Care Medicine, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No.-1-1, Zhongfu Road, Nanjing, 210003, China.
| |
Collapse
|
49
|
Łotowska JM, Sobaniec-Łotowska ME, Bobrus-Chociej A, Sobaniec P. The Ultrastructure of Hepatic Stellate Cell-Macrophage Intercellular Crosstalk as a New Morphological Insight into Phenomenon of Fibrogenesis in Pediatric Autoimmune Hepatitis. J Clin Med 2023; 12:jcm12031024. [PMID: 36769672 PMCID: PMC9917971 DOI: 10.3390/jcm12031024] [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: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
The aim of the study was the pioneering retrospective ultrastructural evaluation of respective forms of hepatic stellate cells (HSCs) and analysis of their crosstalk with other adjacent nonparenchymal cells (NPCs), especially Kupffer cells/macrophages (KCs/MPs), in pediatric autoimmune hepatitis (AIH). METHODS Ultrastructural assessment of the HSC population and NPCs was performed in transmission electron microscopy (TEM) using pretreatment liver biopsies from 25 children (8 boys and 17 girls) aged 4-17 with clinic-pathologically diagnosed untreated AIH. RESULTS Submicroscopic evaluation allowed easy identification of numerous HSCs in the form of transitory cells, i.e., T-HSCs, accompanied by signs of fibrosis. T-HSCs included cells with features of activation initiation (iHSCs) and activation perpetuation (pHSCs), indicating high HSC activation plasticity. The pHSCs were markedly elongated and mainly showed a distinct loss of lipid cytoplasmic material, expanded and dilated channels of granular endoplasmic reticulum, and linear bundles of microfilaments beneath the cell membrane. They were surrounded by usually mature collagen fibers. Frequently activated KCs/MPs adhered directly to T-HSCs. Between them, tight intercellular junctions were formed by means of point desmosomes. CONCLUSIONS Our qualitative TEM observations indicate a key role of T-HSCs in liver fibrogenesis in pediatric AIH, with the essential involvement of activated KCs/MPs that directly adhere to them. Tight intercellular junctions, being the ultrastructural exponent of the specific cellular mechanisms of the crosstalk between NPCs, can play a vital role in hepatic collagen fibroplasia. A better understanding of HSC population morphology at the ultrastructural level in AIH seems important not only to improve the disease morphological diagnostics but to also provide new insights into therapeutic interventions for the phenomenon of liver fibrogenesis.
Collapse
Affiliation(s)
- Joanna Maria Łotowska
- Department of Medical Pathomorphology, Faculty of Medicine with the Division of Dentistry and Division of Medical Education in English, Medical University of Bialystok, 15-269 Bialystok, Poland
- Correspondence: (J.M.Ł.); (P.S.)
| | - Maria Elżbieta Sobaniec-Łotowska
- Department of Medical Pathomorphology, Faculty of Medicine with the Division of Dentistry and Division of Medical Education in English, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Anna Bobrus-Chociej
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Faculty of Medicine with the Division of Dentistry and Division of Medical Education in English, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Piotr Sobaniec
- Department of Pediatric Neurology, Faculty of Medicine with the Division of Dentistry and Division of Medical Education in English, Medical University of Bialystok, 15-274 Bialystok, Poland
- Correspondence: (J.M.Ł.); (P.S.)
| |
Collapse
|
50
|
Akl MG, Widenmaier SB. Immunometabolic factors contributing to obesity-linked hepatocellular carcinoma. Front Cell Dev Biol 2023; 10:1089124. [PMID: 36712976 PMCID: PMC9877434 DOI: 10.3389/fcell.2022.1089124] [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: 11/03/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major public health concern that is promoted by obesity and associated liver complications. Onset and progression of HCC in obesity is a multifactorial process involving complex interactions between the metabolic and immune system, in which chronic liver damage resulting from metabolic and inflammatory insults trigger carcinogenesis-promoting gene mutations and tumor metabolism. Moreover, cell growth and proliferation of the cancerous cell, after initiation, requires interactions between various immunological and metabolic pathways that provide stress defense of the cancer cell as well as strategic cell death escape mechanisms. The heterogenic nature of HCC in addition to the various metabolic risk factors underlying HCC development have led researchers to focus on examining metabolic pathways that may contribute to HCC development. In obesity-linked HCC, oncogene-induced modifications and metabolic pathways have been identified to support anabolic demands of the growing HCC cells and combat the concomitant cell stress, coinciding with altered utilization of signaling pathways and metabolic fuels involved in glucose metabolism, macromolecule synthesis, stress defense, and redox homeostasis. In this review, we discuss metabolic insults that can underlie the transition from steatosis to steatohepatitis and from steatohepatitis to HCC as well as aberrantly regulated immunometabolic pathways that enable cancer cells to survive and proliferate in the tumor microenvironment. We also discuss therapeutic modalities targeted at HCC prevention and regression. A full understanding of HCC-associated immunometabolic changes in obesity may contribute to clinical treatments that effectively target cancer metabolism.
Collapse
Affiliation(s)
- May G. Akl
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Physiology, University of Alexandria, Alexandria, Egypt
| | - Scott B. Widenmaier
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|