1
|
Tang B, Jin C, Li M, Liu S, Zhang X, Li J, Ding K, Zang Y. A novel pectin-like polysaccharide from Crocus sativus targets Galectin-3 to inhibit hepatic stellate cells activation and liver fibrosis. Carbohydr Polym 2025; 348:122826. [PMID: 39562101 DOI: 10.1016/j.carbpol.2024.122826] [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: 04/15/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 11/21/2024]
Abstract
Liver fibrosis may lead to cirrhosis and even cancer without effective clinical medicine available. Previous studies demonstrated that galactan-containing pectins or pectin-like polysaccharides might target Galectin-3 (Gal-3) to impede fibrosis. This research aims to discover novel pectin-like galactan to interfere with fibrosis for potential new drug development. Thus, we purify a novel homogeneous Rhamnogalacturonan-I like polysaccharide with galactan input, XHH2, from the Crocus sativus flower. XHH2 (MW: 35.7 kDa) consists of rhamnose, galacturonic acid, galactose, and arabinose in ratios of 6.6: 6.1: 25.2: 12.1. The backbone of XHH2 comprises 1, 3, 6-Gal and 1, 3, 4-GalA, with branches at O-3 of 1, 3, 6-Gal and O-4 of 1, 3, 4-GalA. O-3 branches include 1, 3-Gal, 1, 4-Gal, 1, 6-Gal, T-Gal, and T-Glc, while O-4 branches consist of 1, 2, 4-Rha, 1, 4-GalA, 1, 5-Ara, T-Ara, T-Gal, and T-α-HexA. Surface plasmon resonance measurement shows that XHH2 binds to both Gal-3 and integrin β1 to block Gal-3/integrin β1 interaction. Mechanism studies further suggest that XHH2 inactivates hepatic stellate cells (HSCs) via disturbing the Gal-3/Integrin-β1/FAK pathway to alleviate liver injury and fibrosis in vitro & in vivo. XHH2 shows a favorable drug safety in the acute toxicity test of oral administration of XHH2 in mice. Overall, XHH2 is an active ingredient against liver fibrosis by targeting the interaction between Gal-3 and Integrin-β1.
Collapse
Affiliation(s)
- Bixi Tang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Can Jin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District; Zhongshan, Guangzhou 528400, China
| | - Maoting Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District; Zhongshan, Guangzhou 528400, China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Siqi Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Jia Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District; Zhongshan, Guangzhou 528400, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang 310024, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Kan Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District; Zhongshan, Guangzhou 528400, China.
| | - Yi Zang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Lingang Laboratory, Shanghai 201203, China.
| |
Collapse
|
2
|
Tang BX, Zhang Y, Sun DD, Liu QY, Li C, Wang PP, Gao LX, Zhang XM, Li J, Zhu WL, Zang Y. Luteolin-7-diglucuronide, a novel PTP1B inhibitor, ameliorates hepatic stellate cell activation and liver fibrosis in mice. Acta Pharmacol Sin 2025; 46:122-133. [PMID: 39103531 PMCID: PMC11697251 DOI: 10.1038/s41401-024-01351-3] [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: 03/29/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024]
Abstract
Liver fibrosis, one of the leading causes of morbidity and mortality worldwide, lacks effective therapy. The activation of hepatic stellate cells (HSCs) is the dominant event in hepatic fibrogenesis. Luteolin-7-diglucuronide (L7DG) is the major flavonoid extracted from Perilla frutescens and Verbena officinalis. Their beneficial effects in the treatment of liver diseases were well documented. In this study we investigated the anti-fibrotic activities of L7DG and the potential mechanisms. We established TGF-β1-activated mouse primary hepatic stellate cells (pHSCs) and human HSC line LX-2 as in vitro liver fibrosis models. Co-treatment with L7DG (5, 20, 50 μM) dose-dependently decreased TGF-β1-induced expression of fibrotic markers collagen 1, α-SMA and fibronectin. In liver fibrosis mouse models induced by CCl4 challenge alone or in combination with HFHC diet, administration of L7DG (40, 150 mg·kg-1·d-1, i.g., for 4 or 8 weeks) dose-dependently attenuated hepatic histopathological injury and collagen accumulation, decreased expression of fibrogenic genes. By conducting target prediction, molecular docking and enzyme activity detection, we identified L7DG as a potent inhibitor of protein tyrosine phosphatase 1B (PTP1B) with an IC50 value of 2.10 µM. Further studies revealed that L7DG inhibited PTP1B activity, up-regulated AMPK phosphorylation and subsequently inhibited HSC activation. This study demonstrates that the phytochemical L7DG may be a potential therapeutic candidate for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Bi-Xi Tang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yong Zhang
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dan-Dan Sun
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qin-Yi Liu
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Cong Li
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Pei-Pei Wang
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li-Xin Gao
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xue-Mei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Jia Li
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| | - Wei-Liang Zhu
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Yi Zang
- Lingang Laboratory, Shanghai, 201203, China.
| |
Collapse
|
3
|
Taga M, Yoshida K, Yano S, Takahashi K, Kyoizumi S, Sasatani M, Suzuki K, Ogawa T, Kusunoki Y, Tsuruyama T. Hepatic Stellate Cell-mediated Increase in CCL5 Chemokine Expression after X-ray Irradiation Determined In Vitro and In Vivo. Radiat Res 2024; 202:862-869. [PMID: 39449628 DOI: 10.1667/rade-23-00127.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Radiation exposure causes hepatitis which induces hepatic steatosis and fibrosis. Although hepatic stellate cells (HSCs) have been considered potential pathological modulators for the development of hepatitis due to viral and microbial infections, their involvement in radiation-induced hepatitis is yet to be determined. This study aimed to clarify the relationship between radiation exposure and expressions of inflammatory cytokines and chemokines in HSCs in vitro and in vivo. HSCs were obtained from 1-week-old mice, known to be highly sensitive to radiation-induced hepatocellular carcinoma, using a newly established method combining liver perfusion, cell dissociation, and density gradient centrifugation, followed by magnetic negative selection of hematopoietic and endothelial cells with anti-CD45.2 and CD146 antibodies. The isolated HSCs were confirmed by the expression of desmin and glial fibrillary acidic protein (GFAP). We demonstrated that primary cultured HSCs, exposed to X-ray irradiation (0, 1.9, and 3.8 Gy) and cultured for 3 and 7 days, produced elevated levels of C-C motif chemokine ligand 5 (CCL5, also known as RANTES) inflammatory chemokine in a dose-dependent manner. An in vivo immunofluorescence method confirmed that increased CCL5 signals were observed in GFAP-positive HSCs in mouse livers 7 days after whole-body X-ray irradiation (1.9 and 3.8 Gy). Adequate expression of C-C motif chemokine receptor 5 (Ccr5), a receptor for CCL5, was also detected using real-time PCR in the liver of both irradiated and non-irradiated mice. Taken together, our data suggest that HSCs may drive hepatitis via CCL5/CCR5 axis in the liver under radiation-induced stress. Furthermore, this newly established experimental protocol can help evaluate the expression of other inflammatory cytokines in primary cultures of HSCs isolated from infant mice.
Collapse
Affiliation(s)
- Masataka Taga
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima City, Hiroshima, Japan
| | - Kengo Yoshida
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima City, Hiroshima, Japan
| | - Shiho Yano
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima City, Hiroshima, Japan
| | - Keiko Takahashi
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima City, Hiroshima, Japan
| | - Seishi Kyoizumi
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima City, Hiroshima, Japan
| | - Megumi Sasatani
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Keiji Suzuki
- Radiation Risk Control Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tomohiro Ogawa
- Center for the Advancement of Higher Education, Faculty of Engineering, Kindai University, Hiroshima, Japan
| | - Yoichiro Kusunoki
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima City, Hiroshima, Japan
| | - Tatsuaki Tsuruyama
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima City, Hiroshima, Japan
- Department of Drug Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
4
|
Yao H, He Q, Xiang L, Liu S, Yang Z, Li X, Liu W, Huang C, Wang B, Xie Q, Gao Y, Zheng C, Li X. Guizhi Fuling Wan attenuates tetrachloromethane-induced hepatic fibrosis in rats via PTEN/AKT/mTOR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118593. [PMID: 39032663 DOI: 10.1016/j.jep.2024.118593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Treatment options for hepatic fibrosis, a prevalent liver condition closely linked to cirrhosis, are currently limited. While Guizhi Fuling Wan (GFW), a pill derived from traditional Chinese herbs, has been reported to possess hepatoprotective properties, its therapeutic effect and mechanism in hepatic fibrosis remain elusive. AIM OF THE STUDY This study aimed to evaluate the anti-fibrotic impact of GFW and its underlying mechanisms in both in vivo and in vitro settings. MATERIALS AND METHODS Tetrachloromethane (CCl4) was used to induce hepatic fibrosis in male rats. In vitro, activation of hepatic stellate cells (HSCs) was triggered by platelet-derived growth factor-BB (PDGF-BB). In vivo, liver function, pathological alterations, and HSC activation were evaluated. Additionally, the impact of GFW on the activated phenotypes of Lieming Xu-2 (LX-2) cells was examined in vitro. Network pharmacology was employed to identify the potential targets of GFW in hepatic fibrosis. Lastly, the impact of GFW on the PTEN/AKT/mTOR pathway and PTEN ubiquitination in HSCs was investigated. RESULTS GFW alleviated CCl4-induced liver damage and scarring in rats in a dose-dependent manner and suppressed HSC activation in vivo. Moreover, GFW inhibited the proliferation, migration, differentiation, and extracellular matrix (ECM) production of activated HSCs in vitro. GFW also promoted autophagy and apoptosis of HSCs. Meanwhile, network pharmacology and in vitro studies suggested that GFW inhibits the AKT/mTOR pathway by preventing PTEN degradation by suppressing ubiquitination. CONCLUSION GFW attenuates Ccl4-induced hepatic fibrosis in male rats by regulating the PTEN/AKT/mTOR signaling pathway, positioning it as a potential candidate for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Huan Yao
- Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Tianfu TCM Innovation Harbour, Chengdu University of Traditional Chinese Medicine, Chengdu 611930, China.
| | - Qingman He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| | - Li Xiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| | - Sixian Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| | - Zhuodi Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| | - Xue Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| | - Weiwei Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Cong Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Baojia Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Qian Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Yongxiang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan, China.
| | - Chuan Zheng
- Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Tianfu TCM Innovation Harbour, Chengdu University of Traditional Chinese Medicine, Chengdu 611930, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Xueping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| |
Collapse
|
5
|
Ma Y, Harris J, Li P, Jiang C, Sun H, Cao H. An Integrative Transcriptome Subtraction Strategy to Identify Human lncRNAs That Specifically Play a Role in Activation of Human Hepatic Stellate Cells. Noncoding RNA 2024; 10:34. [PMID: 38921831 PMCID: PMC11206700 DOI: 10.3390/ncrna10030034] [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: 03/06/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Abstract
Fibrotic liver features excessive deposition of extracellular matrix (ECM), primarily produced from "activated" hepatic stellate cells (HSCs). While targeting human HSCs (hHSCs) in fibrosis therapeutics shows promise, the overall understanding of hHSC activation remains limited, in part because it is very challenging to define the role of human long non-coding RNAs (lncRNAs) in hHSC activation. To address this challenge, we identified another cell type that acts via a diverse gene network to promote fibrogenesis. Then, we identified the lncRNAs that were differentially regulated in activated hHSCs and the other profibrotic cell. Next, we conducted concurrent analysis to identify those lncRNAs that were specifically involved in fibrogenesis. We tested and confirmed that transdifferentiation of vascular smooth muscle cells (VSMCs) represents such a process. By overlapping TGFβ-regulated lncRNAs in multiple sets of hHSCs and VSMCs, we identified a highly selected list of lncRNA candidates that could specifically play a role in hHSC activation. We experimentally characterized one human lncRNA, named CARMN, which was significantly regulated by TGFβ in all conditions above. CARMN knockdown significantly reduced the expression levels of a panel of marker genes for hHSC activation, as well as the levels of ECM deposition and hHSC migration. Conversely, gain of function of CARMN using CRISPR activation (CRISPR-a) yielded the completely opposite effects. Taken together, our work addresses a bottleneck in identifying human lncRNAs that specifically play a role in hHSC activation and provides a framework to effectively select human lncRNAs with significant pathophysiological role.
Collapse
Affiliation(s)
| | | | | | | | | | - Haiming Cao
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Hu Q, Liu Z, Liu Y, Qiu J, Zhang X, Sun J, Zhang B, Shi H. SIAH2 suppresses c-JUN pathway by promoting the polyubiquitination and degradation of HBx in hepatocellular carcinoma. J Cell Mol Med 2024; 28:e18484. [PMID: 38842124 PMCID: PMC11154841 DOI: 10.1111/jcmm.18484] [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: 02/02/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024] Open
Abstract
As an important protein encoded by hepatitis B virus (HBV), HBV X protein (HBx) plays an important role in the development of hepatocellular carcinoma (HCC). It has been shown that seven in absentia homologue 1 (SIAH1) could regulates the degradation of HBx through the ubiquitin-proteasome pathway. However, as a member of SIAH family, the regulatory effects of SIAH2 on HBx remain unclear. In this study, we first confirmed that SIAH2 could reduce the protein levels of HBx depending on its E3 ligase activity. Moreover, SIAH2 interacted with HBx and induced its K48-linked polyubiquitination and proteasomal degradation. Furthermore, we provided evidence that SIAH2 inhibits HBx-associated HCC cells proliferation by regulating HBx. In conclusion, our study identified a novel role for SIAH2 in promoting HBx degradation and SIAH2 exerts an inhibitory effect in the proliferation of HBx-associated HCC through inducing the degradation of HBx. Our study provides a new idea for the targeted degradation of HBx and may have great huge significance into providing novel evidence for the targeted therapy of HBV-infected HCC.
Collapse
Affiliation(s)
- Qinghe Hu
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Zhiyi Liu
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Yao Liu
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Jie Qiu
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Xue Zhang
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Jun Sun
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Bin Zhang
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| | - Hengliang Shi
- Institute of Digestive DiseasesXuzhou Medical UniversityXuzhouJiangsuChina
- Research Center of Digestive DiseasesThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of General SurgeryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Central LaboratoryThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
| |
Collapse
|
7
|
Raymant M, Astuti Y, Alvaro-Espinosa L, Green D, Quaranta V, Bellomo G, Glenn M, Chandran-Gorner V, Palmer DH, Halloran C, Ghaneh P, Henderson NC, Morton JP, Valiente M, Mielgo A, Schmid MC. Macrophage-fibroblast JAK/STAT dependent crosstalk promotes liver metastatic outgrowth in pancreatic cancer. Nat Commun 2024; 15:3593. [PMID: 38678021 PMCID: PMC11055860 DOI: 10.1038/s41467-024-47949-3] [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: 07/10/2023] [Accepted: 04/16/2024] [Indexed: 04/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease for which better therapies are urgently needed. Fibroblasts and macrophages are heterogeneous cell populations able to enhance metastasis, but the role of a macrophage-fibroblast crosstalk in regulating their pro-metastatic functions remains poorly understood. Here we deconvolve how macrophages regulate metastasis-associated fibroblast (MAF) heterogeneity in the liver. We identify three functionally distinct MAF populations, among which the generation of pro-metastatic and immunoregulatory myofibroblastic-MAFs (myMAFs) critically depends on macrophages. Mechanistically, myMAFs are induced through a STAT3-dependent mechanism driven by macrophage-derived progranulin and cancer cell-secreted leukaemia inhibitory factor (LIF). In a reciprocal manner, myMAF secreted osteopontin promotes an immunosuppressive macrophage phenotype resulting in the inhibition of cytotoxic T cell functions. Pharmacological blockade of STAT3 or myMAF-specific genetic depletion of STAT3 restores an anti-tumour immune response and reduces metastases. Our findings provide molecular insights into the complex macrophage-fibroblast interactions in tumours and reveal potential targets to inhibit PDAC liver metastasis.
Collapse
Affiliation(s)
- Meirion Raymant
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Yuliana Astuti
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Laura Alvaro-Espinosa
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Daniel Green
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Valeria Quaranta
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Gaia Bellomo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Mark Glenn
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Vatshala Chandran-Gorner
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Daniel H Palmer
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Christopher Halloran
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Paula Ghaneh
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jennifer P Morton
- Cancer Research-UK Scotland Institute and School of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow, G61 1BD, UK
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
8
|
Ren Y, Chen Y, Tang EH, Hu Y, Niu B, Liang H, Xi C, Zhao F, Cao Z. Arbidol attenuates liver fibrosis and activation of hepatic stellate cells by blocking TGF-β1 signaling. Eur J Pharmacol 2024; 967:176367. [PMID: 38325795 DOI: 10.1016/j.ejphar.2024.176367] [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: 11/02/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
Chronic liver diseases (CLD) impact over 800 million people globally, causing about 2 million deaths annually. Arbidol (ARB), an indole-derivative used to treat influenza virus infection, was extensively used during COVID-19 pandemic in China. In recent years, studies have shown that ARB, compared to other antiviral drugs, exhibits greater liver-protective efficacy, indicating a potential hepatoprotective effect beyond its antiviral activity. However, the mechanism remains unclear. In this study, we investigated the impact of ARB on liver injury/fibrosis in bile duct ligated (BDL) mice and its effect on spontaneous and transforming growth factor β1 (TGF-β1)-induced activation of primary cultured hepatic stellate cells (HSCs). Oral administration of ARB significantly ameliorated BDL-induced liver injury/fibrosis as reflected by decreased serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), reduced collagen deposition, and diminished mRNA expression of fibrosis markers. ARB notably inhibited spontaneous and TGF-β1-induced activation of primary cultured HSCs. Moreover, ARB also drastically attenuated mRNA expression levels of platelet-derived growth factor receptor (Pdgfr), transforming growth factor-beta receptor (Tgfbr) 1, Tgfbr2, matrix metalloproteinase (Mmp)-2, and Mmp-9 in activated HSCs. We further demonstrate that ARB mitigated Smad2/3 phosphorylation in both TGF-β1 treated HSCs and BDL mice. These data together demonstrate that the therapeutic efficacy of ARB on liver fibrosis is independent of its antiviral activity and likely is achieved by blocking TGF-β1 signaling-mediated HSC activation.
Collapse
Affiliation(s)
- Younan Ren
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ying Chen
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Emily H Tang
- BASIS International School Nanjing, No.18 Lingshan North Road, Qixia District, Nanjing, Jiangsu, 210023, China
| | - Yixin Hu
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China; Animal Experiment Center of China Pharmaceutical University, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Bo Niu
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Huaduan Liang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Chuchu Xi
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Fang Zhao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
9
|
Tsuchiya Y, Seki T, Kobayashi K, Komazawa-Sakon S, Shichino S, Nishina T, Fukuhara K, Ikejima K, Nagai H, Igarashi Y, Ueha S, Oikawa A, Tsurusaki S, Yamazaki S, Nishiyama C, Mikami T, Yagita H, Okumura K, Kido T, Miyajima A, Matsushima K, Imasaka M, Araki K, Imamura T, Ohmuraya M, Tanaka M, Nakano H. Fibroblast growth factor 18 stimulates the proliferation of hepatic stellate cells, thereby inducing liver fibrosis. Nat Commun 2023; 14:6304. [PMID: 37813881 PMCID: PMC10562492 DOI: 10.1038/s41467-023-42058-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/28/2023] [Indexed: 10/11/2023] Open
Abstract
Liver fibrosis results from chronic liver injury triggered by factors such as viral infection, excess alcohol intake, and lipid accumulation. However, the mechanisms underlying liver fibrosis are not fully understood. Here, we demonstrate that the expression of fibroblast growth factor 18 (Fgf18) is elevated in mouse livers following the induction of chronic liver fibrosis models. Deletion of Fgf18 in hepatocytes attenuates liver fibrosis; conversely, overexpression of Fgf18 promotes liver fibrosis. Single-cell RNA sequencing reveals that overexpression of Fgf18 in hepatocytes results in an increase in the number of Lrat+ hepatic stellate cells (HSCs), thereby inducing fibrosis. Mechanistically, FGF18 stimulates the proliferation of HSCs by inducing the expression of Ccnd1. Moreover, the expression of FGF18 is correlated with the expression of profibrotic genes, such as COL1A1 and ACTA2, in human liver biopsy samples. Thus, FGF18 promotes liver fibrosis and could serve as a therapeutic target to treat liver fibrosis.
Collapse
Affiliation(s)
- Yuichi Tsuchiya
- Department of Biochemistry, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama, Funabashi-shi, Chiba, 274-8510, Japan
| | - Takao Seki
- Department of Biochemistry, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Kenta Kobayashi
- Department of Biochemistry, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Sachiko Komazawa-Sakon
- Department of Biochemistry, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda-shi, Chiba, 278-0022, Japan
| | - Takashi Nishina
- Department of Biochemistry, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Kyoko Fukuhara
- Department of Gastroenterology, Faculty of Medicine and Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Kenichi Ikejima
- Department of Gastroenterology, Faculty of Medicine and Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Hidenari Nagai
- Department of Gastroenterology, Toho University Omori Medical Center, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Yoshinori Igarashi
- Department of Gastroenterology, Toho University Omori Medical Center, 6-11-1 Omori-Nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda-shi, Chiba, 278-0022, Japan
| | - Akira Oikawa
- Laboratory of Quality Analysis and Assessment, Graduate School of Agriculture, Kyoto University, Gokasyo, Uji-shi, Kyoto, 611-0011, Japan
| | - Shinya Tsurusaki
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Tokyo, Japan
- Laboratory of Stem Cell Regulation, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Tokyo, Japan
| | - Soh Yamazaki
- Department of Biochemistry, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Chiharu Nishiyama
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan
| | - Tetuo Mikami
- Department of Pathology, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Hideo Yagita
- Department of Immunology, Faculty of Medicine and Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Ko Okumura
- Atopy Research Center, Faculty of Medicine and Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Taketomo Kido
- Laboratory of Cell Growth and Differentiation, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Atsushi Miyajima
- Laboratory of Cell Growth and Differentiation, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda-shi, Chiba, 278-0022, Japan
| | - Mai Imasaka
- Department of Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya-shi, Hyogo, 663-8501, Japan
| | - Kimi Araki
- Center for Animal Resources and Development, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Toru Imamura
- Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
- National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Umezono, Tsukuba-shi, Ibaraki, 305-8560, Japan
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya-shi, Hyogo, 663-8501, Japan
| | - Minoru Tanaka
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Tokyo, Japan
- Laboratory of Stem Cell Regulation, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Tokyo, Japan
| | - Hiroyasu Nakano
- Department of Biochemistry, Faculty of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan.
| |
Collapse
|
10
|
Lang Z, Li Y, Lin L, Li X, Tao Q, Hu Y, Bao M, Zheng L, Yu Z, Zheng J. Hepatocyte-derived exosomal miR-146a-5p inhibits hepatic stellate cell EMT process: a crosstalk between hepatocytes and hepatic stellate cells. Cell Death Discov 2023; 9:304. [PMID: 37598186 PMCID: PMC10439924 DOI: 10.1038/s41420-023-01602-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
Recently, Salidroside (Sal) has been demonstrated to suppress hepatic stellate cell (HSC) activation, a crucial event for liver fibrosis. Moreover, Sal has been reported to decrease hepatocyte injury. A growing number of reports have indicated that the crosstalk between hepatocytes and HSCs is very crucial for liver fibrosis development. Whether Sal-treated hepatocytes could inhibit HSC activation is unclear. Exosomes, as vital vehicles of intercellular communication, have been shown to transfer cargos between hepatocytes and HSCs. Herein, we aimed to investigate the roles of exosomal miRNAs from Sal-treated hepatocytes in HSC activation as well as liver fibrosis. Our results showed that Sal suppressed carbon tetrachloride (CCl4)-induced liver fibrosis in vivo. HSC activation as well as cell proliferation was repressed in HSCs co-cultured with Sal-treated hepatocytes. Interestingly, miR-146a-5p was up-regulated by Sal in CCl4-treated mice. Also, enhanced miR-146a-5p was found in hepatocytes isolated from Sal-treated CCl4 mice and hepatocyte-derived exosomes. Notably, hepatocyte exosomal miR-146a-5p contributed to HSC inactivation. Inhibiting miR-146a-5p in hepatocyte exosomes resulted in reduced E-cadherin (E-cad) and increased desmin in HSCs, indicating that miR-146a-5p caused HSC inactivation via epithelial-mesenchymal transition (EMT). miR-146a-5p inhibition-mediated HSC activation and EMT process were blocked down by loss of EIF5A2. Further studies revealed that EIF5A2 was a target of miR-146a-5p. Furthermore, exosomes with miR-146a-5p overexpression inhibited liver fibrosis in CCl4 mice. Collectively, exosomal miR-146a-5p from Sal-treated hepatocytes inhibits HSC activation and liver fibrosis, at least in part, by suppressing EIF5A2 and EMT process.
Collapse
Affiliation(s)
- Zhichao Lang
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yifei Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lifan Lin
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xinmiao Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qiqi Tao
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuhang Hu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315000, China
| | - Menglu Bao
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lei Zheng
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhengping Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jianjian Zheng
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315000, China.
| |
Collapse
|
11
|
Qi J, Ping D, Sun X, Huang K, Peng Y, Liu C. A herbal product inhibits carbon tetrachloride-induced liver fibrosis by suppressing the epidermal growth factor receptor signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116419. [PMID: 37003405 DOI: 10.1016/j.jep.2023.116419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzheng Huayu formula (FZHY), composed of Salvia miltiorrhiza Bunge, Cordyceps sinensis, the seed of Prunus persica (L.) Batsch, the pollen of Pinus massoniana Lamb, Gynostemma pentaphyllum (Thunb.) Makino and the fruit of Schisandra chinensis (Turcz.) Baill, is a Chinese herbal compound with demonstrated clinical benefits in liver fibrosis (LF). However, its potential mechanism and molecular targets remain to be elucidated. AIM OF THE STUDY This study was designed to evaluate the anti-fibrotic role of FZHY in hepatic fibrosis and to elucidate the potential mechanisms. MATERIALS AND METHODS Network pharmacology was assayed to identify the interrelationships among compounds of FZHY, potential targets and putative pathways on anti-LF. Then the core pharmaceutical target for FZHY against LF was verified by serum proteomic analysis. Further in vivo and in vitro assays were performed to verify the prediction of the pharmaceutical network. RESULTS The network pharmacology analysis revealed that a total of 175 FZHY-LF crossover proteins were filtered into a protein-protein interaction (PPI) network complex and designated as the potential targets of FZHY against LF, and the Epidermal Growth Factor Receptor (EGFR) signaling pathway was further explored according to the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Then analytical studies were validated by carbon tetrachloride (CCl4)-induced model in vivo. We found FZHY could attenuate CCl4-induced LF, especially decrease p-EGFR expression in α-Smooth Muscle Actin (α-SMA)-positive hepatic stellate cell (HSC) and inhibit the downstream of the EGFR signaling pathway, especially Extracellular Regulated Protein Kinases (ERK) signaling pathway in liver tissue. We further demonstrate that FZHY could inhibit Epidermal Growth Factor (EGF)-induced HSC activation, as well as the expression of p-EGFR and the key protein of the ERK signaling pathway. CONCLUSIONS FZHY has a good effect against CCl4-induced LF. The action mechanism was associated with the down-regulation of the EGFR signaling pathway in activated HSCs.
Collapse
Affiliation(s)
- Jingshu Qi
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dabing Ping
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Sun
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Kai Huang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan Peng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China; Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai, 201203, China.
| |
Collapse
|
12
|
Lang Z, Yu S, Hu Y, Tao Q, Zhang J, Wang H, Zheng L, Yu Z, Zheng J. Ginsenoside Rh2 promotes hepatic stellate cell ferroptosis and inactivation via regulation of IRF1-inhibited SLC7A11. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154950. [PMID: 37441987 DOI: 10.1016/j.phymed.2023.154950] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/13/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Sustained liver fibrosis may lead to cirrhosis. Activated hepatic stellate cells (HSCs) are crucial for liver fibrosis development. Ferroptosis, a newly iron-dependent regulated cell death, has been demonstrated to be involved in HSC inactivation. PURPOSE Ginsenoside Rh2 (GRh2), a natural bioactive product derived from ginseng, has been shown to promote HSC inactivation. However, the effect of GRh2 on HSC ferroptosis remains unclear. METHODS We explored the effects of GRh2 on liver fibrosis in vivo and in vitro. RNA-sequence analysis was performed in HSCs after GRh2 treatment. The crosstalk between ferroptotic HSCs and macrophages was also explored. RESULTS GRh2 alleviated liver fibrosis in vivo. In vitro, GRh2 reduced HSC proliferation and activation via ferroptosis, with increased intracellular iron, reactive oxygen species, malondialdehyde and glutathione depletion. The expression of SLC7A11, a negative regulator of ferroptosis, was obviously reduced by GRh2. Interestingly, interferon regulatory factor 1 (IRF1), a transcription factor, was predicted to bind the promoter region of SCL7A11. The interaction between IRF1 and SCL7A11 was further confirmed by the results of chromatin immunoprecipitation and luciferase reporter assays. Furthermore, loss of IRF1 led to an increase in SCL7A11, which contributed to the suppression of HSC ferroptosis and the enhancement of HSC activation in GRh2-treated HSCs. Further studies revealed that GRh2-induced HSC ferroptosis contributed to the inhibition of macrophage recruitment via regulation of inflammation-related genes. Moreover, GRh2 caused a reduction in liver inflammation in vivo. CONCLUSION Collectively, GRh2 up-regulates IRF1 expression, resulting in the suppression of SLC7A11, which contributes to HSC ferroptosis and inactivation. GRh2 ameliorates liver fibrosis through enhancing HSC ferroptosis and inhibiting liver inflammation. GRh2 may be a promising drug for treating liver fibrosis.
Collapse
Affiliation(s)
- Zhichao Lang
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Suhui Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yuhang Hu
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qiqi Tao
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jingnan Zhang
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Haoyue Wang
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lei Zheng
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhixian Yu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Jianjian Zheng
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
13
|
Torres S, Ortiz C, Bachtler N, Gu W, Grünewald LD, Kraus N, Schierwagen R, Hieber C, Meier C, Tyc O, Joseph Brol M, Uschner FE, Nijmeijer B, Welsch C, Berres M, Garcia‐Ruiz C, Fernandez‐Checa JC, Trautwein C, Vogl TJ, Zeuzem S, Trebicka J, Klein S. Janus kinase 2 inhibition by pacritinib as potential therapeutic target for liver fibrosis. Hepatology 2023; 77:1228-1240. [PMID: 35993369 PMCID: PMC10026969 DOI: 10.1002/hep.32746] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Janus kinase 2 (JAK2) signaling is increased in human and experimental liver fibrosis with portal hypertension. JAK2 inhibitors, such as pacritinib, are already in advanced clinical development for other indications and might also be effective in liver fibrosis. Here, we investigated the antifibrotic role of the JAK2 inhibitor pacritinib on activated hepatic stellate cells (HSCs) in vitro and in two animal models of liver fibrosis in vivo . APPROACH AND RESULTS Transcriptome analyses of JAK2 in human livers and other targets of pacritinib have been shown to correlate with profibrotic factors. Although transcription of JAK2 correlated significantly with type I collagen expression and other profibrotic genes, no correlation was observed for interleukin-1 receptor-associated kinase and colony-stimulating factor 1 receptor. Pacritinib decreased gene expression of fibrosis markers in mouse primary and human-derived HSCs in vitro . Moreover, pacritinib decreased the proliferation, contraction, and migration of HSCs. C 57 BL/6J mice received ethanol in drinking water (16%) or Western diet in combination with carbon tetrachloride intoxication for 7 weeks to induce alcoholic or nonalcoholic fatty liver disease. Pacritinib significantly reduced liver fibrosis assessed by gene expression and Sirius red staining, as well as HSC activation assessed by alpha-smooth muscle actin immunostaining in fibrotic mice. Furthermore, pacritinib decreased the gene expression of hepatic steatosis markers in experimental alcoholic liver disease. Additionally, pacritinib protected against liver injury as assessed by aminotransferase levels. CONCLUSIONS This study demonstrates that the JAK2 inhibitor pacritinib may be promising for the treatment of alcoholic and nonalcoholic liver fibrosis and may be therefore relevant for human pathology.
Collapse
Affiliation(s)
- Sandra Torres
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Liver Unit‐IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Cristina Ortiz
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Nadine Bachtler
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Wenyi Gu
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Leon D. Grünewald
- Department of Diagnostic and Interventional Radiology, Universit+y Hospital Frankfurt, Frankfurt am Main, Germany
| | - Nico Kraus
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Robert Schierwagen
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Christoph Hieber
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Caroline Meier
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Olaf Tyc
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Maximilian Joseph Brol
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Frank Erhard Uschner
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Bart Nijmeijer
- Research and Development Department, Linxis BV, Amsterdam, The Netherlands
| | - Christoph Welsch
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Marie‐Luise Berres
- Department of Internal Medicine III, Aachen University Hospital, Aachen, Germany
| | - Carmen Garcia‐Ruiz
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Liver Unit‐IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Department of Medicine, University of Southern California, Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jose Carlos Fernandez‐Checa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Liver Unit‐IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Department of Medicine, University of Southern California, Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Christian Trautwein
- Department of Internal Medicine III, Aachen University Hospital, Aachen, Germany
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional Radiology, Universit+y Hospital Frankfurt, Frankfurt am Main, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
- European Foundation for the Study of Chronic Liver Failure – EF Clif, Barcelona, Spain
| | - Sabine Klein
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| |
Collapse
|
14
|
Wang W, Zhang Y, Jiang Y, Wang Y, Zhu J, Wang C, Han X, Wang J. Exploration of potential mechanism of Rougan formula against hepatic fibrosis by network analysis and experimental assessment. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:115960. [PMID: 36565772 DOI: 10.1016/j.jep.2022.115960] [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: 09/21/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rougan Formula (RG) has long been clinically applied to treat hepatic fibrosis in patients with different chronic liver diseases. However, the core active substances and the potential pharmacological mechanisms of RG remain unclear. AIM OF THE STUDY The purpose of this study is to explore bioactive components, key targets, and potential mechanisms of RG by performing network pharmacological analyses and experimental model validation. MATERIALS AND METHODS All chemical components in RG extract were identified using ultraperformance liquid chromatography-quadrupole/time-of-flight tandem mass technology. The candidate components and drug targets of RG, as well as disease-related genes, were extracted from TCMSP and GeneCards databases. The potential pathways related to genes were predicted by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. The core bioactive components, key targets, and signaling pathways were ultimately obtained by analyzing protein-protein interaction (PPI) and component-target-pathway (C-T-P) networks. Subsequently, the efficacy and underlying mechanisms of RG on hepatic fibrosis were experimentally validated in transforming growth factor-beta 1 (TGF-β1)-induced hepatic stellate cell activation model and CCL4-induced hepatic fibrosis mouse model. RESULTS A total of 52 components in RG extract were obtained, and 22 of them were selected as the core bioactive components. Five hundred and thirty-nine overlapped targets were determined by matching drug targets with disease-related targets. The results of PPI and C-T-P network analyses revealed 100 key targets and 19 signaling pathways associated with RG efficacy. In vitro and in vivo studies further verified that RG exerted a significant anti-hepatic fibrotic effect by suppressing the activation of hepatic stellate cells by downregulating the TGF-β1/Smads signaling pathway. CONCLUSIONS These results may provide some evidence for further clinical research and development of RG formula as an effective and safe drug for hepatic fibrosis treatment.
Collapse
Affiliation(s)
- Wenyi Wang
- Department of Liver Disease, Shanghai Yueyang Integrated Traditional Chinese Medicine and Western Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yu Zhang
- Department of Liver Disease, Shanghai Yueyang Integrated Traditional Chinese Medicine and Western Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yue Jiang
- Engineering Research Center of Modernization of Traditional Chinese Medicine, East China University of Science and Technology, Shanghai, China.
| | - Yujie Wang
- Department of Liver Disease, Shanghai Yueyang Integrated Traditional Chinese Medicine and Western Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Junfeng Zhu
- Department of Liver Disease, Shanghai Yueyang Integrated Traditional Chinese Medicine and Western Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Chunli Wang
- Engineering Research Center of Modernization of Traditional Chinese Medicine, East China University of Science and Technology, Shanghai, China.
| | - Xianghui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jianyi Wang
- Department of Liver Disease, Shanghai Yueyang Integrated Traditional Chinese Medicine and Western Medicine Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
15
|
Schröder SK, Tag CG, Weiskirchen S, Weiskirchen R. Phalloidin Staining for F-Actin in Hepatic Stellate Cells. Methods Mol Biol 2023; 2669:55-66. [PMID: 37247054 DOI: 10.1007/978-1-0716-3207-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
During the development of liver fibrosis, hepatic stellate cells undergo a transition from a quiescent phenotype into a proliferative, fibrogenic, and contractile, α-smooth muscle actin-positive myofibroblast. These cells acquire properties that are strongly associated with the reorganization of the actin cytoskeleton. Actin possesses a unique ability to polymerize into filamentous actin (F-actin) form its monomeric globular state (G-actin). F-actin can form robust actin bundles and cytoskeletal networks by interacting with a number of actin-binding proteins that provide important mechanical and structural support for a multitude of cellular processes including intracellular transport, cell motility, polarity, cell shape, gene regulation, and signal transduction. Therefore, stains with actin-specific antibodies and phalloidin conjugates for actin staining are widely used to visualize actin structures in myofibroblasts. Here we present an optimized protocol for F-actin staining for hepatic stellate cells using a fluorescent phalloidin.
Collapse
Affiliation(s)
- Sarah K Schröder
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany.
| | - Carmen G Tag
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institut für Molekulare Pathobiochemie, Experimentelle Gentherapie und Klinische Chemie (IFMPEGKC), Universitätsklinikum Aachen AöR, Aachen, Germany.
| |
Collapse
|
16
|
Meurer SK, Weiskirchen S, Tag CG, Weiskirchen R. Isolation, Purification, and Culture of Primary Murine Hepatic Stellate Cells: An Update. Methods Mol Biol 2023; 2669:1-32. [PMID: 37247051 DOI: 10.1007/978-1-0716-3207-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
In the healthy liver, quiescent hepatic stellate cells (HSCs) are found in the perisinusoidal space (i.e., the space of Dissé) in close proximity to endothelial cells and hepatocytes. HSCs represent 5-8% of the total number of liver cells and are characterized by numerous fat vacuoles that store vitamin A in the form of retinyl esters. Upon liver injury caused by different etiologies, HSCs become activated and acquire a myofibroblast (MFB) phenotype in a process called transdifferentiation. In contrast to quiescent HSC, MFB become highly proliferative and are characterized by an imbalance in extracellular matrix (ECM) homeostasis, by producing an excess of collagen and blocking its turnover by synthesis of protease inhibitors. This leads to a net accumulation of ECM during fibrosis. In addition to HSC, there are fibroblasts in the portal fields (pF), which also have the potency to acquire a myofibroblastic phenotype (pMF). The contributions of these two fibrogenic cell types (i.e., MFB and pMF) vary based on the etiology of liver damage (parenchymal vs. cholestatic). Based on their importance to hepatic fibrosis, the isolation and purification protocols of these primary cells are in great demand. Moreover, established cell lines may offer only limited information about the in vivo behavior of HSC/MFB and pF/pMF.Here we describe a method for high-purity isolation of HSC from mice. In the first step, the liver is digested with pronase and collagenase, and the cells are dissociated from the tissue. In the second step, HSCs are enriched by density gradient centrifugation of the crude cell suspension using a Nycodenz gradient. The resulting cell fraction can be further optionally purified by flow cytometric enrichment to generate ultrapure HSC.
Collapse
Affiliation(s)
- Steffen K Meurer
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany.
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany.
| | - Carmen G Tag
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- IInstitut für Molekulare Pathobiochemie, Experimentelle Gentherapie und Klinische Chemie (IFMPEGKC), Universitätsklinikum Aachen AöR, Aachen, Germany
| |
Collapse
|
17
|
Fan L, Pan Q, Yang W, Koo SC, Tian C, Li L, Lu M, Brown A, Ju B, Easton J, Ranganathan S, Shin S, Bondoc A, Yang JJ, Yu J, Zhu L. A developmentally prometastatic niche to hepatoblastoma in neonatal liver mediated by the Cxcl1/Cxcr2 axis. Hepatology 2022; 76:1275-1290. [PMID: 35179799 PMCID: PMC9385889 DOI: 10.1002/hep.32412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Hepatoblastoma (HB) is the most common pediatric liver cancer. Its predominant occurrence in very young children led us to investigate whether the neonatal liver provides a protumorigenic niche to HB development. APPROACH AND RESULTS HB development was compared between orthotopic transplantation models established in postnatal day 5 (P5) and 60 (P60) mice (P5Tx and P60Tx models). Single-cell RNA-sequencing (sc-RNAseq) was performed using tumor and liver tissues from both models and the top candidate cell types and genes identified are investigated for their roles in HB cell growth, migration, and survival. CONCLUSIONS We found that various HB cell lines including HepG2 cells were consistently and considerably more tumorigenic and metastatic in the P5Tx model than in the P60Tx models. Sc-RNAseq of the P5Tx and P60Tx HepG2 models revealed that the P5Tx tumor was more hypoxic and had a larger number of activated hepatic stellate cells (aHSCs) in the tumor-surrounding liver that express significantly higher levels of Cxcl1 than those from the P60Tx model. We found these differences were developmentally present in normal P5 and P60 liver. We showed that the Cxcl1/Cxcr2 axis mediated HB cell migration and was critical to HB cell survival under hypoxia. Treating HepG2 P60Tx model with recombinant CXCL1 protein induced intrahepatic and pulmonary metastasis and CXCR2 knockout (KO) in HepG2 cells abolished their metastatic potential in the P5Tx model. Lastly, we showed that in tumors from patients with metastatic HB, there was a similar larger population of aHSCs in the tumor-surrounding liver than in localized tumors, and tumor hypoxia was uniquely associated with prognosis of patients with HB among pediatric cancers. We demonstrated that the neonatal liver provides a prometastatic niche to HB development through the Cxcl1/Cxcr2 axis.
Collapse
Affiliation(s)
- Li Fan
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Qingfei Pan
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Wentao Yang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Selene C. Koo
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Cheng Tian
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Liyuan Li
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Meifen Lu
- Veterinary Pathology Core, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Anthony Brown
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Bensheng Ju
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - John Easton
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Sarangarajan Ranganathan
- Department of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital, Cincinnati, Ohio, United States
| | - Soona Shin
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Alexander Bondoc
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Jun J. Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Liqin Zhu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| |
Collapse
|
18
|
Yan FZ, Qian H, Liu F, Ding CH, Liu SQ, Xiao MC, Chen SJ, Zhang X, Luo C, Xie WF. Inhibition of protein arginine methyltransferase 1 alleviates liver fibrosis by attenuating the activation of hepatic stellate cells in mice. FASEB J 2022; 36:e22489. [PMID: 35959865 DOI: 10.1096/fj.202200238r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 11/11/2022]
Abstract
Protein arginine methyltransferase 1 (PRMT1) has been reported to be involved in various diseases. The expression of PRMT1 was increased in cirrhotic livers from human patients. However, the role of PRMT1 in hepatic fibrogenesis remains largely unexplored. In this study, we investigated the effect of PRMT1 on hepatic fibrogenesis and its underlying mechanism. We found that PRMT1 expression was significantly higher in fibrotic livers of the mice treated with thioacetamide (TAA) or 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet. Immunofluorescence staining revealed that PRMT1 expression was augmented in both hepatocytes and hepatic stellate cells (HSCs) in the fibrotic livers. Applying a selective inhibitor of PRMT1, PT1001B, significantly suppressed PRMT1 activity and mitigated liver fibrosis in mice. Hepatocyte-specific Prmt1 knockout did not affect liver fibrosis in mice. PRMT1 overexpression promoted the expression of fibrotic genes in the LX-2 cells, whereas knockdown of PRMT1 or treatment with PT1001B exhibited reversal effects, suggesting that PRMT1 plays an important role in HSC activation. Additionally, HSC-specific Prmt1 knockout attenuated HSC activation and liver fibrosis in TAA-induced fibrotic model. RNA-seq analysis revealed that Prmt1 knockout in HSCs significantly suppressed pro-inflammatory NF-κB and pro-fibrotic TGF-β signals, and also downregulated the expression of pro-fibrotic mediators in mouse livers. Moreover, treatment with PT1001B consistently inhibited hepatic inflammatory response in fibrotic model. In conclusion, PRMT1 plays a vital role in HSC activation. Inhibition of PRMT1 mitigates hepatic fibrosis by attenuating HSC activation in mice. Therefore, targeting PRMT1 could be a feasible therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Fang-Zhi Yan
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Hui Qian
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fang Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen-Hong Ding
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shu-Qing Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Meng-Chao Xiao
- Department of Gastroenterology, Shanghai East Hospital, Tongji University of School of Medicine, Shanghai, China
| | - Shi-Jie Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, The Center for Chemical Biology, Drug Discovery and Design Center, Chinese Academy of Sciences, Shanghai, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Cheng Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, The Center for Chemical Biology, Drug Discovery and Design Center, Chinese Academy of Sciences, Shanghai, China
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
19
|
Hydrophobic Bile Salts Induce Pro-Fibrogenic Proliferation of Hepatic Stellate Cells through PI3K p110 Alpha Signaling. Cells 2022; 11:cells11152344. [PMID: 35954188 PMCID: PMC9367387 DOI: 10.3390/cells11152344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/08/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Bile salts accumulating during cholestatic liver disease are believed to promote liver fibrosis. We have recently shown that chenodeoxycholate (CDC) induces expansion of hepatic stellate cells (HSCs) in vivo, thereby promoting liver fibrosis. Mechanisms underlying bile salt-induced fibrogenesis remain elusive. We aimed to characterize the effects of different bile salts on HSC biology and investigated underlying signaling pathways. Murine HSCs (mHSCs) were stimulated with hydrophilic and hydrophobic bile salts. Proliferation, cell mass, collagen deposition, and activation of signaling pathways were determined. Activation of the human HSC cell line LX 2 was assessed by quantification of α-smooth muscle actin (αSMA) expression. Phosphatidyl-inositol-3-kinase (PI3K)-dependent signaling was inhibited both pharmacologically and by siRNA. CDC, the most abundant bile salt accumulating in human cholestasis, but no other bile salt tested, induced Protein kinase B (PKB) phosphorylation and promoted HSC proliferation and subsequent collagen deposition. Pharmacological inhibition of the upstream target PI3K-inhibited activation of PKB and pro-fibrogenic proliferation of HSCs. The PI3K p110α-specific inhibitor Alpelisib and siRNA-mediated knockdown of p110α ameliorated pro-fibrogenic activation of mHSC and LX 2 cells, respectively. In summary, pro-fibrogenic signaling in mHSCs is selectively induced by CDC. PI3K p110α may be a potential therapeutic target for the inhibition of bile salt-induced fibrogenesis in cholestasis.
Collapse
|
20
|
Li Y, Wei M, Yuan Q, Liu Y, Tian T, Hou L, Zhang J. MyD88 in hepatic stellate cells promotes the development of alcoholic fatty liver via the AKT pathway. J Mol Med (Berl) 2022; 100:1071-1085. [PMID: 35708745 DOI: 10.1007/s00109-022-02196-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
Myeloid differentiation primary response gene 88 (MyD88), an adaptor protein in the Toll-like receptors (TLRs) signalling pathway, is expressed in various liver cells including hepatocytes, Kupffer cells and hepatic stellate cells (HSCs). And yet, the functional role of MyD88 in HSCs is poorly elucidated in alcoholic fatty liver (AFL). Here, to study the functional role of MyD88 in HSCs and the molecular mechanism related to the development of AFL, chronic-binge ethanol mouse models were established in mice with specific MyD88 knockout in quiescent (MyD88GFAP-KO) and activated HSCs (MyD88SMA-KO), respectively. Our results clearly showed an elevated expression of MyD88 in liver tissues of ethanol treated mouse model which harbours the wild type. Intriguingly, ethanol treatment profoundly inhibited inflammation in both MyD88GFAP-KO and MyD88SMA-KO mice, but the suppression of lipogenesis was only observed in MyD88GFAP-KO mice. Molecularly, our study indicated that MyD88 induced osteopontin (OPN) secretion in HSCs, which consequently resulted in activation of AKT signalling pathway and accumulation of fat in hepatocytes. Additionally, our data also suggested that OPN promoted inflammation by activating p-STAT1. Thus, targeting MyD88 may be a potentially represent a promising strategy for the prevention and treatment of AFL. KEY MESSAGES: The expression of MyD88 in HSCs was significantly increased in ethanol-induced liver tissues of wild-type mice. MyD88 deficiency in quiescent HSCs inhibited inflammation and lipogenesis under the ethanol feeding condition. MyD88 deficiency in activated HSCs only inhibited inflammation under the ethanol feeding condition. MyD88 promoted the OPN secretion of HSCs, which further activated the AKT signalling pathway of hepatocytes and upregulated lipogenic gene expression to promote fat accumulation. OPN also promotes inflammation by activating p-STAT1.
Collapse
Affiliation(s)
- Yukun Li
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, People's Republic of China
| | - Miaomiao Wei
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, People's Republic of China
| | - Qi Yuan
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, People's Republic of China
| | - Yu Liu
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, People's Republic of China
| | - Tian Tian
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, People's Republic of China
| | - Lingling Hou
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, People's Republic of China.
| | - Jinhua Zhang
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, People's Republic of China.
| |
Collapse
|
21
|
Bi X, Yang N, Ke Y, Liu H, Ma W, Fang B, Sun L, Li L, Lü G, Lin R. An improved experimental method for simultaneously isolating hepatocytes and hepatic stellate cells in mouse liver infected with Echinococcus multilocularis. J Clin Lab Anal 2021; 35:e24084. [PMID: 34724252 PMCID: PMC8649337 DOI: 10.1002/jcla.24084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 09/24/2021] [Accepted: 10/20/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Alveolar echinococcosis (AE) is a zoonotic disease caused by the larval stage of Echinococcus multilocularis parasitizing in the human liver, causing local pathological changes in the liver and manifesting as hyperplasia, liver fibrosis, atrophy, degeneration, and necrosis. Here, we report a method that can simultaneously isolate hepatocytes and hepatic stellate cells (HSCs) from mice infected with Echinococcus multilocularis. METHODS A mouse model of AE was established. Hepatocytes and HSCs were isolated from mouse liver using a two-step method combining in situ collagenase perfusion and gradient centrifugation. Expressions of Alb, Desmin, and α-SMA were detected with immunofluorescence to identify the isolated hepatocytes and HSCs. RESULTS The viability and purity of hepatocytes and HSCs both reached 90% or above. For hepatocytes, clear cell boundaries were observed, and the nuclei were round or oval, with clear nucleoli. There was a homogeneous distribution of the hepatocyte marker Alb in the cytoplasm of hepatocytes. Lipid droplets and Desmin expression were observed in the cytoplasm of freshly isolated HSCs. During the activation of HSCs, the lipid droplets gradually decreased and disappeared with a high expression of α-SMA. CONCLUSION Hepatocytes and HSCs are simultaneously isolated. This may provide a research tool to investigate the interaction between hepatocytes and HSCs and to investigate the mechanism of Echinococcus multilocularis infection-induced liver pathological changes.
Collapse
Affiliation(s)
- Xiaojuan Bi
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Ning Yang
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Ying Ke
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Hui Liu
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Wenmei Ma
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Bingbing Fang
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Li Sun
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Liang Li
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
| | - Guodong Lü
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
- Department of Epidemiology and Health StatisticsSchool of Public HealthXinjiang Medical UniversityUrumqiChina
| | - Renyong Lin
- State Key Laboratory of PathogenesisPrevention, and Treatment of Central Asian High Incidence DiseasesClinical Medical Research InstituteThe First Affiliated Hospital of Xinjiang Medical UniversityUrumqiChina
- Basic Medical CollegeXinjiang Medical UniversityUrumqiChina
| |
Collapse
|
22
|
Benitez R, Caro M, Andres-Leon E, O'Valle F, Delgado M. CORTISTATIN REGULATES FIBROSIS AND MYOFIBROBLAST ACTIVATION IN EXPERIMENTAL HEPATOTOXIC- AND CHOLESTATIC-INDUCED LIVER INJURY. Br J Pharmacol 2021; 179:2275-2296. [PMID: 34821378 DOI: 10.1111/bph.15752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/08/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Liver fibrosis induced by chronic hepatic injury remains as a major cause of morbidity and mortality worldwide. Identification of susceptibility/prognosis factors and new therapeutic tools for treating hepatic fibrotic disorders are urgent medical needs. Cortistatin is a neuropeptide with potent anti-inflammatory and anti-fibrotic activities in lung that binds to receptors that are expressed in liver fibroblasts and hepatic stellate cells. We evaluated the capacity of cortistatin to regulate liver fibrosis. EXPERIMENTAL APPROACH We experimentally induced liver fibrosis in mice by chronic CCl4 exposition and bile duct ligation and evaluated the histopathological signs and fibrotic markers. KEY RESULTS Hepatic expression of cortistatin inversely correlated with liver fibrosis grade in mice and humans with hepatic disorders. Cortistatin-deficient mice showed exacerbated signs of liver damage and fibrosis and increased mortality rates when challenged to hepatotoxic and cholestatic injury. Compared to wild-type mice, non-parenchymal liver cells isolated from cortistatin-deficient mice showed increased presence of cells with activated myofibroblast phenotypes and a differential genetic signature that is indicative of activated hepatic stellate cells and periportal fibroblasts and of myofibroblasts with active contractile apparatus. Cortistatin treatment reversed in vivo and in vitro these exaggerated fibrogenic phenotypes and protected from progression to severe liver fibrosis in response to hepatic injury. CONCLUSION AND IMPLICATIONS We identify cortistatin as an endogenous molecular break of liver fibrosis and its deficiency as a potential poor-prognosis marker for chronic hepatic disorders that course with fibrosis. Cortistatin-based therapies emerge as attractive strategies for ameliorating severe hepatic fibrosis of various etiologies.
Collapse
Affiliation(s)
- Raquel Benitez
- Institute of Parasitology and Biomedicine Lopez-Neyra, PT Salud, Granada, Spain
| | - Marta Caro
- Institute of Parasitology and Biomedicine Lopez-Neyra, PT Salud, Granada, Spain
| | - Eduardo Andres-Leon
- Institute of Parasitology and Biomedicine Lopez-Neyra, PT Salud, Granada, Spain
| | - Francisco O'Valle
- Dept. of Pathology, School of Medicine, IBIMER and IBS-Granada, University of Granada, Spain
| | - Mario Delgado
- Institute of Parasitology and Biomedicine Lopez-Neyra, PT Salud, Granada, Spain
| |
Collapse
|
23
|
Foresight regarding drug candidates acting on the succinate-GPR91 signalling pathway for non-alcoholic steatohepatitis (NASH) treatment. Biomed Pharmacother 2021; 144:112298. [PMID: 34649219 DOI: 10.1016/j.biopha.2021.112298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, and it is a liver manifestation of metabolic syndrome, with a histological spectrum from simple steatosis to non-alcoholic steatohepatitis (NASH). NASH can evolve into progressive liver fibrosis and eventually lead to liver cirrhosis. The pathological mechanism of NASH is multifactorial, involving a series of metabolic disorders and changes that trigger low-level inflammation in the liver and other organs. In the pathogenesis of NASH, the signal transduction pathway involving succinate and the succinate receptor (G-protein-coupled receptor 91, GPR91) regulates inflammatory cell activation and liver fibrosis. This review describes the mechanism of the succinate-GPR91 signalling pathway in NASH and summarizes the drugs that act on this pathway, with the aim of providing a new approach to NASH treatment.
Collapse
|
24
|
Perilipin 5 Ameliorates Hepatic Stellate Cell Activation via SMAD2/3 and SNAIL Signaling Pathways and Suppresses STAT3 Activation. Cells 2021; 10:cells10092184. [PMID: 34571833 PMCID: PMC8467115 DOI: 10.3390/cells10092184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/12/2022] Open
Abstract
Comprehending the molecular mechanisms underlying hepatic fibrogenesis is essential to the development of treatment. The hallmark of hepatic fibrosis is the development and deposition of excess fibrous connective tissue forcing tissue remodeling. Hepatic stellate cells (HSC) play a major role in the pathogenesis of liver fibrosis. Their activation via the transforming growth factor-β1 (TGF-β1) as a key mediator is considered the crucial event in the pathophysiology of hepatic fibrogenesis. It has been shown that Perilipin 5 (PLIN5), known as a lipid droplet structural protein that is highly expressed in oxidative tissue, can inhibit such activation through various mechanisms associated with lipid metabolism. This study aimed to investigate the possible influence of PLIN5 on TGF-β1 signaling. Our findings confirm the importance of PLIN5 in maintaining HSC quiescence in vivo and in vitro. PLIN5 overexpression suppresses the TGF-β1-SMAD2/3 and SNAIL signaling pathways as well as the activation of the signal transducers and activators of transcription 3 (STAT3). These findings derived from experiments in hepatic cell lines LX-2 and Col-GFP, in which overexpression of PLIN5 was able to downregulate the signaling pathways SMAD2/3 and SNAIL activated previously by TGF-β1 treatment. Furthermore, TGF-β1-mediatedinduction of extracellular matrix proteins, such as collagen type I (COL1), Fibronectin, and α-smooth muscle actin (α-SMA), was suppressed by PLIN5. Moreover, STAT3, which is interrelated with TGF-β1 was already basally activated in the cell lines and inhibited by PLIN5 overexpression, leading to a further reduction in HSC activity shown by lowered α-SMA expression. This extension of the intervening mechanisms presents PLIN5 as a potent and pleiotropic target in HSC activation.
Collapse
|
25
|
McCabe MC, Schmitt LR, Hill RC, Dzieciatkowska M, Maslanka M, Daamen WF, van Kuppevelt TH, Hof DJ, Hansen KC. Evaluation and Refinement of Sample Preparation Methods for Extracellular Matrix Proteome Coverage. Mol Cell Proteomics 2021; 20:100079. [PMID: 33845168 PMCID: PMC8188056 DOI: 10.1016/j.mcpro.2021.100079] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
The extracellular matrix is a key component of tissues, yet it is underrepresented in proteomic datasets. Identification and evaluation of proteins in the extracellular matrix (ECM) has proved challenging due to the insolubility of many ECM proteins in traditional protein extraction buffers. Here we separate the decellularization and ECM extraction steps of several prominent methods for evaluation under real-world conditions. The results are used to optimize a two-fraction ECM extraction method. Approximately one dozen additional parameters are tested, and recommendations for analysis based on overall ECM coverage or specific ECM classes are given. Compared with a standard in-solution digest, the optimized method yielded a fourfold improvement in unique ECM peptide identifications.
Collapse
Affiliation(s)
- Maxwell C McCabe
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Ryan C Hill
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, USA; Cancer Center Proteomics Core, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, USA; Cancer Center Proteomics Core, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Mark Maslanka
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, USA; Cancer Center Proteomics Core, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Willeke F Daamen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Danique J Hof
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, USA; Cancer Center Proteomics Core, School of Medicine, University of Colorado, Aurora, Colorado, USA.
| |
Collapse
|
26
|
Tang Y, Ma N, Luo H, Chen S, Yu F. Downregulated long non-coding RNA LINC01093 in liver fibrosis promotes hepatocyte apoptosis via increasing ubiquitination of SIRT1. J Biochem 2021; 167:525-534. [PMID: 32044992 DOI: 10.1093/jb/mvaa013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/25/2019] [Indexed: 02/07/2023] Open
Abstract
The apoptosis of hepatocytes contributes to the activation of hepatic stellate cells (HSCs), thus promoting the accumulation of extracellular matrix proteins and aggravating liver fibrosis. Silent information regulator 1 (SIRT1) is an anti-fibrotic protein whose downregulation induces hepatocyte apoptosis. This study aims to identify whether SIRT1 is regulated by long non-coding RNA LINC01093 and explore its underlying mechanisms. Liver fibrosis was induced in mice using CCl4, and the differential expressions of several fibrosis-related long noncoding RNAs were detected in liver tissues. The effect of LINC01093 on cell apoptosis and viability of hepatocytes were investigated after LINC01093 overexpression or knockdown using flow cytometry and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. The anti-fibrotic effect of LINC01093 overexpression was observed in vivo. LncRNA LINC01093 is downregulated in CCl4-induced liver tissues and TGF-β1-stimulated hepatocytes. Downregulated LINC01093 promoted cell apoptosis and inhibited cell viability of hepatocytes. The co-culture between LINC01093-knockdown hepatocytes and HSCs increased the expressions of pro-fibrotic proteins. Downregulated LINC01093 promoted hepatocyte apoptosis via promoting degradation and ubiquitination of SIRT1 under TGF-β1 stimulation. The injection of LINC01093-overexpressing vectors alleviated liver fibrosis in vivo. In liver fibrosis, the downregulated LINC01093 promoted hepatocyte apoptosis, which is mediated by increasing the degradation and ubiquitination of SIRT1.
Collapse
Affiliation(s)
- Yinhe Tang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, 205 Wenrui Avenue, Wenzhou 325000, People's Republic of China
| | - Naijing Ma
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, 205 Wenrui Avenue, Wenzhou 325000, People's Republic of China
| | - Hao Luo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, 205 Wenrui Avenue, Wenzhou 325000, People's Republic of China
| | - Shizuan Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, 205 Wenrui Avenue, Wenzhou 325000, People's Republic of China
| | - Fuxiang Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, 205 Wenrui Avenue, Wenzhou 325000, People's Republic of China
| |
Collapse
|
27
|
Dang TM, Le T, Do H, Nguyen V, Holterman A, Dang LT, Phan NC, Pham PV, Hoang S, Le L, Grassi G, Truong N. Optimization of the isolation procedure and culturing conditions for hepatic stellate cells obtained from mouse. Biosci Rep 2021; 41:BSR20202514. [PMID: 33350435 PMCID: PMC7823195 DOI: 10.1042/bsr20202514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/28/2020] [Accepted: 12/18/2020] [Indexed: 12/28/2022] Open
Abstract
Liver fibrosis (LF) mortality rate is approximately 2 million per year. Irrespective of the etiology of LF, a key element in its development is the transition of hepatic stellate cells (HSCs) from a quiescent phenotype to a myofibroblast-like cell with the production of fibrotic proteins. It is necessary to define optimal isolation and culturing conditions for good HSCs yield and proper phenotype preservation for studying the activation of HSCs in vitro. In the present study, the optimal conditions of HSC isolation and culture were examined to maintain the HSC's undifferentiated phenotype. HSCs were isolated from Balb/c mice liver using Nycodenz, 8, 9.6, and 11%. The efficiency of the isolation procedure was evaluated by cell counting and purity determination by flow cytometry. Quiescent HSCs were cultured in test media supplemented with different combinations of fetal bovine serum (FBS), glutamine (GLN), vitamin A (vitA), insulin, and glucose. The cells were assessed at days 3 and 7 of culture by evaluating the morphology, proliferation using cell counting kit-8, lipid storage using Oil Red O (ORO) staining, expression of a-smooth muscle actin, collagen I, and lecithin-retinol acyltransferase by qRT-PCR and immunocytochemistry (ICC). The results showed that Nycodenz, at 9.6%, yielded the best purity and quantity of HSCs. Maintenance of HSC undifferentiated phenotype was achieved optimizing culturing conditions (serum-free Dulbecco's Modified Eagle's Medium (DMEM) supplemented with glucose (100 mg/dl), GLN (0.5 mM), vitA (100 μM), and insulin (50 ng/ml)) with a certain degree of proliferation allowing their perpetuation in culture. In conclusion, we have defined optimal conditions for HSCs isolation and culture.
Collapse
Affiliation(s)
- Thanh Minh Dang
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
- School of Biotechnology, International University, VNUHCM, Ho Chi Minh City, Vietnam
| | - Trinh Van Le
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
| | - Huy Quang Do
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
| | - Van Thuan Nguyen
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
- School of Biotechnology, International University, VNUHCM, Ho Chi Minh City, Vietnam
| | - Ai Xuan Le Holterman
- Department of Pediatrics and Surgery, University of Illinois College of Medicine, Chicago and Peoria, Illinois, U.S.A
| | - Loan Tung Thi Dang
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
- Faculty of Biology and Biotechnology, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
| | - Nhan Chinh Lu Phan
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
| | - Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
| | - Son Nghia Hoang
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Long Thanh Le
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Trieste, Italy
| | - Nhung Hai Truong
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
- University of Science, Viet Nam National University, Ho Chi Minh City, Vietnam
- Faculty of Biology and Biotechnology, University of Science, VNUHCM, Ho Chi Minh City, Vietnam
| |
Collapse
|
28
|
Zhang Z, Wen H, Peng B, Weng J, Zeng F. Downregulated microRNA-129-5p by Long Non-coding RNA NEAT1 Upregulates PEG3 Expression to Aggravate Non-alcoholic Steatohepatitis. Front Genet 2021; 11:563265. [PMID: 33574830 PMCID: PMC7870803 DOI: 10.3389/fgene.2020.563265] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have recently emerged as inflammation-associated biological molecules with a specific role in the progression of liver fibrosis conditions including non-alcoholic steatohepatitis (NASH). The aim of this study was to elucidate the effects of lncRNA nuclear enriched abundant transcript 1 (NEAT1), microRNA-129-5p (miR-129-5p), and paternally expressed gene 3 (PEG3) on the biological activities of hepatic stellate cells (HSCs) subjected to NASH. First, microarray-based analysis revealed upregulated PEG3 in NASH. Liver tissues from mice fed a methionine–choline-deficient (MCD) diet exhibited increased expression of NEAT1 and PEG3 along with lower miR-129-5p expression. A series of in vitro and in vivo assays were then performed on HSCs after transfection with shPEG3, miR-129-5p mimic, or treatment with pyrrolidine dithiocarbamate (PDTC), an inhibitor of the nuclear factor-kappa B (NF-κB) signaling pathway. Results confirmed the alleviated fibrosis by restoring miR-129-5p, while depleting PEG3 or NEAT1, as evidenced by the inactivation of HSCs. To sum up, NEAT1 can bind specifically to miR-129-5p and consequently regulate miR-129-5p and PEG3 expression in relation to the HSC activation occurring in NASH. Thus, NEAT1-targeted inhibition against miR-129-5p presents a promising therapeutic strategy for the treatment of NASH.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Huiqing Wen
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Bangjian Peng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Jun Weng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fanhong Zeng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
29
|
Wang K, Wang J, Song M, Wang H, Xia N, Zhang Y. Angelica sinensis polysaccharide attenuates CCl 4-induced liver fibrosis via the IL-22/STAT3 pathway. Int J Biol Macromol 2020; 162:273-283. [PMID: 32569681 DOI: 10.1016/j.ijbiomac.2020.06.166] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022]
Abstract
Angelica sinensis polysaccharide (ASP) has hepatoprotective effects in liver injury models. However, its role and mechanism in chronic liver fibrosis have not been fully elucidated. In this study, a carbon tetrachloride (CCl4)-induced chronic liver fibrosis mouse model was established. The results showed that ASP treatment reduced serum alanine aminotransferase by approximately 50% and liver fibrosis areas by approximately 70%. Hepatic stellate cell (HSC) activation was inhibited in ASP-treated mice. Furthermore, the mechanism was studied in-depth, focusing on the interleukin 22/signal transducer and activator of transcription 3 (IL-22/STAT3) axis. Concentrations of 50 μg/ml and 100 μg/ml ASP induced the secretion of IL-22 in vitro, which further increased at a concentration of 200 μg/ml. Moreover, in vivo data showed that ASP significantly promoted IL-22 production in splenocytes and liver tissues. The antifibrotic effects of ASP were abolished after IL-22 neutralization. In addition, ASP activated the STAT3 pathway in the liver, as demonstrated by a 2-fold increase compared to that of the CCl4 group, which was abrogated by the IL-22 antibody. Subsequently, we showed that the antifibrotic effects of ASP were abrogated by blocking STAT3 with S3I-201. In conclusion, ASP effectively alleviates chronic liver fibrosis by inhibiting HSC activation through the IL-22/STAT3 pathway.
Collapse
Affiliation(s)
- Kaiping Wang
- Hubei Key Laboratory of Nature Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Junfeng Wang
- Hubei Key Laboratory of Nature Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Mengzi Song
- Hubei Key Laboratory of Nature Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Hanxiang Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
30
|
Wu P, Luo X, Wu H, Zhang Q, Dai Y, Sun M. Efficient and targeted chemo-gene delivery with self-assembled fluoro-nanoparticles for liver fibrosis therapy and recurrence. Biomaterials 2020; 261:120311. [PMID: 32911091 DOI: 10.1016/j.biomaterials.2020.120311] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022]
Abstract
The treatment options of liver fibrosis remain limited except for liver transplantation due to the complexity and slow development in its progression. Besides, liver fibrosis recurrence and intervention time have not been reported as significant indicators to affect the anti-fibrotic efficacy of tested drugs/strategies. Herein, a novel fluoropolymer is developed to achieve high drug loading of sorafenib and efficient delivery of miR155 inhibitor (anti-miR155) for dual-targeting of hepatic stellate cells (HSCs) and kupffer cells (KCs), and we report a detailed plan on the design of treatment regimen to reveal the relationship between chemogene therapy, intervention time and fibrosis recurrence. Such a combined chemo-gene therapy of sorafenib and anti-miR155 can achieve superior therapeutic efficiency by polarizing the pro-inflammatory M1 to anti-inflammatory M2 of KCs and inhibiting the proliferation of HSCs. Importantly, efficacy and recurrence prevention of chemogene therapy earlier in the liver fibrosis will be more effective than the treatment at later stage. In conclusion, this work proposes a novel strategy to improve the efficacy and prevent recurrence of liver fibrosis by dual-regulating of KCs and HSCs, and emphasizes the importance of therapy earlier in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Pengkai Wu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Xinping Luo
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Hui Wu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Qingyan Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuanxin Dai
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
31
|
Ramachandran P, Matchett KP, Dobie R, Wilson-Kanamori JR, Henderson NC. Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis. Nat Rev Gastroenterol Hepatol 2020; 17:457-472. [PMID: 32483353 DOI: 10.1038/s41575-020-0304-x] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/08/2020] [Indexed: 12/19/2022]
Abstract
Liver disease is a major global health-care problem, affecting an estimated 844 million people worldwide. Despite this substantial burden, therapeutic options for liver disease remain limited, in part owing to a paucity of detailed analyses defining the cellular and molecular mechanisms that drive these conditions in humans. Single-cell transcriptomic technologies are transforming our understanding of cellular diversity and function in health and disease. In this Review, we discuss how these technologies have been applied in hepatology, advancing our understanding of cellular heterogeneity and providing novel insights into fundamental liver biology such as the metabolic zonation of hepatocytes, endothelial cells and hepatic stellate cells, and the cellular mechanisms underpinning liver regeneration. Application of these methodologies is also uncovering critical pathophysiological changes driving disease states such as hepatic fibrosis, where distinct populations of macrophages, endothelial cells and mesenchymal cells reside within a spatially distinct fibrotic niche and interact to promote scar formation. In addition, single-cell approaches are starting to dissect key cellular and molecular functions in liver cancer. In the near future, new techniques such as spatial transcriptomics and multiomic approaches will further deepen our understanding of disease pathogenesis, enabling the identification of novel therapeutic targets for patients across the spectrum of liver diseases.
Collapse
Affiliation(s)
- Prakash Ramachandran
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Kylie P Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - John R Wilson-Kanamori
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK. .,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
32
|
Yin L, Zhang Y, Shi H, Feng Y, Zhang Z, Zhang L. Proteomic profiling of hepatic stellate cells in alcohol liver fibrosis reveals proteins involved in collagen production. Alcohol 2020; 86:81-91. [PMID: 32171770 DOI: 10.1016/j.alcohol.2020.02.167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 01/17/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hepatic stellate cell (HSC) activation has central functions in alcohol-induced liver fibrosis. Proteins of HSCs in alcoholic liver fibrosis (ALF) are still not completely understood. Here, we performed a proteomic study to discover proteins related to ALF using HSCs isolated from a rat model. METHODS Sprague-Dawley rats were fed with ethanol for 2 or 6 weeks. Liver histology was assessed using Sirius red and Oil red O staining. HSCs were enriched by using Percoll density gradient centrifugation, and analyzed using flow cytometry. Proteins extracted from HSCs were separated using two-dimensional electrophoresis (2DE). Differentially expressed proteins were identified using liquid chromatography-mass spectrometry (LC-MS). The characteristics of the differentially expressed proteins were analyzed using the UniProtKB database and STRING software. The mRNA levels of two differentially expressed proteins were analyzed using real-time RT-PCR, of which NADH dehydrogenase (ubiquinone) flavoprotein 2, mitochondrial (Ndufv2) was further investigated using Western blot (WB) and immunohistochemical analysis in the ALF model and human liver tissues. The relationship between Ndufv2 and alcohol stimulation was evaluated using WB. Next, Ndufv2 was knocked-down by shRNA in the HSC-T6 cell line. Three genes (encoding collagen, metalloproteinase inhibitor 1 [TIMP-1], and α-smooth muscle actin [a-SMA]) related to HSC activation were detected. RESULTS An ALF model was successfully established, with a liver fibrosis score of 1-2 (S1-2), and some big fat vacuoles development. Twenty-one non-abundant proteins with more than a 2-fold difference were identified using mass spectrometry, including 7 upregulated and 14 downregulated proteins. These differential proteins are a response to antigen presentation, mitochondrial metabolism, ethanol, and collagen degradation. Among them, two upregulated proteins (Ndufv2 and ATP synthase subunit alpha, mitochondrial [ATP5a1]) were involved in mitochondrial metabolism in ALF, and showed concurrent changes in mRNA and protein levels. Ndufv2 was upregulated in HSCs, as shown by WB, in non-parenchymal cells (NPCs) in the rat model and human liver tissues, and detected using immunohistochemistry. Ndufv2 was also upregulated after alcohol stimulation. Following Ndufv2 knockdown, collagen, TIMP-1, and α-SMA were downregulated compared with that in the controls. CONCLUSIONS A proteomic study was performed to discover proteins related to ALF in HSCs isolated from a rat model. Twenty-one differentially expressed proteins were identified, including proteins involved in mitochondrial metabolism and antigen presentation. Ndufv2, an upregulated protein in ALF, might be involved in ALF through regulating the production of fibrosis factors.
Collapse
|
33
|
|
34
|
Li S, Song F, Lei X, Li J, Li F, Tan H. hsa_circ_0004018 suppresses the progression of liver fibrosis through regulating the hsa-miR-660-3p/TEP1 axis. Aging (Albany NY) 2020; 12:11517-11529. [PMID: 32584784 PMCID: PMC7343491 DOI: 10.18632/aging.103257] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 04/17/2020] [Indexed: 02/07/2023]
Abstract
Efforts have been made in the prevention and treatment of liver fibrosis. The inhibition or depletion of the hepatic stellate cells (HSCs) has been considered as a potential approach. Recently, there are numbers of studies about the role of the circular RNA in the disease progression. However, the role of circular RNA in the regulation of HSCs and the progression of liver fibrosis remained elusive. In this study, we constructed a CCl4-induced liver fibrosis mouse model and overexpressed hsa_circ_0004018 in HSCs. Then, salvianolic acid B was used to treat HSCs in vitro. We found that hsa_circ_0004018 is downregulated in liver fibrogenesis. Luciferase reporter assay was performed to verify the interaction of hsa_circ_0004018, hsa-miR-660-3p and TEP1. It showed that hsa_circ_0004018 may act as a sponge of hsa-miR-660-3p, which can target and downregulate the expression of TEP1. hsa_circ_0004018 expressing lentivirus was used to investigate the in-vivo function of hsa_circ_0004018 in CCl4-induced liver fibrosis mice. We also reveal that the hsa_circ_0004018/hsa-miR-660-3p/TEP1 axis contributes to the proliferation and activation of HSCs. In addition, the overexpression of hsa_circ_0004018 alleviated the progression of liver fibrosis. In conclusion, our study highlights hsa_circ_0004018 as a potential biomarker and therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Shan Li
- Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Fangmin Song
- Department of Infectious Diseases, People's Hospital of Yunxi, Shiyan, Hubei, China
| | - Xu Lei
- Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jingtao Li
- Department of Liver Diseases, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Fang Li
- Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Huabing Tan
- Department of Infectious Diseases and Lab of Liver Disease, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
35
|
Li Y, Zhang Y, Chen T, Huang Y, Zhang Y, Geng S, Li X. Role of aldosterone in the activation of primary mice hepatic stellate cell and liver fibrosis via NLRP3 inflammasome. J Gastroenterol Hepatol 2020; 35:1069-1077. [PMID: 31860730 DOI: 10.1111/jgh.14961] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/19/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Emerging evidence suggests aldosterone (aldo) and NLRP3 inflammasome are important factors for HSC activation and liver fibrosis. However, the interaction between aldo and NLRP3 inflammasome in HSC activation and liver fibrosis remains largely unknown. The aim of this study is to investigate the relationship between aldo and NLRP3 inflammasome in liver fibrosis. METHODS Serum and liver specimens collected from 40 patients with or without liver fibrosis were used to test the level of aldo and NLRP3. Primary HSC isolated from C57BL/6 mice were treated with aldo, and the effects of aldo on NLRP3 inflammasome and HSC activation were detected in vitro. Two animal models were used to verify the effect of aldo on liver fibrosis in vivo: hyperaldosteronism model was established in wild-type and NLRP3 knockout (NLRP3-/- ) mice by micro-pump, and liver fibrosis mouse model was built by tetrachloromethane (CCl4 ). RESULTS Patients with liver fibrosis showed higher aldo levels and increased NLRP3 expression in liver. In vitro, aldo induced the activation of primary mouse HSCs by promoting the expression and assembly of NLRP3 inflammasome. In vivo, NLRP3 knockout could alleviate the liver fibrosis induced by aldo in mice. In addition, treatment with spironolactone (spi) could inhibit the NLRP3 expression, HSC activation, and liver fibrosis induced by CCl4 . CONCLUSIONS Aldo promotes the activation of HSCs and liver fibrosis through NLRP3 inflammasome relative pathways. Intervention of aldo and NLRP3 inflammasome-related pathways may provide a promising strategy for treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yijie Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yun Huang
- Department of Cadre's Ward, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yan Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shiyu Geng
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
36
|
Hübbers A, Hennings J, Lambertz D, Haas U, Trautwein C, Nevzorova YA, Sonntag R, Liedtke C. Pharmacological Inhibition of Cyclin-Dependent Kinases Triggers Anti-Fibrotic Effects in Hepatic Stellate Cells In Vitro. Int J Mol Sci 2020; 21:ijms21093267. [PMID: 32380742 PMCID: PMC7246535 DOI: 10.3390/ijms21093267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 04/28/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a wound healing process in response to chronic liver injury, which is characterized by the accumulation of extracellular collagen produced by Hepatic Stellate Cells (HSCs). This process involves cell cycle re-entry and proliferation of normally quiescent HSCs controlled by cyclins and associated cyclin-dependent kinases (Cdks). Cdk2 mediates the entry and progression through S-phase in complex with E-and A-type cyclins. We have demonstrated that cyclin E1 is essential for liver fibrogenesis in mice, but it is not known if this is dependent on Cdk2 or related Cdks. Here, we aimed to evaluate the benefit of the pan-Cdk inhibitor CR8 for treatment of liver fibrosis in vitro. CR8-treatment reduced proliferation and survival in immortalized HSC lines and in addition attenuated pro-fibrotic properties in primary murine HSCs. Importantly, primary murine hepatocytes were much more tolerant against the cytotoxic and anti-proliferative effects of CR8. We identified CR8 dosages mediating anti-fibrotic effects in primary HSCs without affecting cell cycle activity and survival in primary hepatocytes. In conclusion, the pharmacological pan-Cdk inhibitor CR8 restricts the pro-fibrotic properties of HSCs, while preserving proliferation and viability of hepatocytes at least in vitro. Therefore, CR8 and related drugs might be beneficial for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Anna Hübbers
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Julia Hennings
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Daniela Lambertz
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Ute Haas
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Yulia A. Nevzorova
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
- Department of Genetics, Physiology, and Microbiology, Faculty of Biology, Complutense University Madrid, 28040 Madrid, Spain
| | - Roland Sonntag
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
- Correspondence: (R.S.); (C.L.)
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
- Correspondence: (R.S.); (C.L.)
| |
Collapse
|
37
|
Recent Advances in Practical Methods for Liver Cell Biology: A Short Overview. Int J Mol Sci 2020; 21:ijms21062027. [PMID: 32188134 PMCID: PMC7139397 DOI: 10.3390/ijms21062027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022] Open
Abstract
Molecular and cellular research modalities for the study of liver pathologies have been tremendously improved over the recent decades. Advanced technologies offer novel opportunities to establish cell isolation techniques with excellent purity, paving the path for 2D and 3D microscopy and high-throughput assays (e.g., bulk or single-cell RNA sequencing). The use of stem cell and organoid research will help to decipher the pathophysiology of liver diseases and the interaction between various parenchymal and non-parenchymal liver cells. Furthermore, sophisticated animal models of liver disease allow for the in vivo assessment of fibrogenesis, portal hypertension and hepatocellular carcinoma (HCC) and for the preclinical testing of therapeutic strategies. The purpose of this review is to portray in detail novel in vitro and in vivo methods for the study of liver cell biology that had been presented at the workshop of the 8th meeting of the European Club for Liver Cell Biology (ECLCB-8) in October of 2018 in Bonn, Germany.
Collapse
|
38
|
Ye L, Yu Y, Zhao Y. Icariin-induced miR-875-5p attenuates epithelial-mesenchymal transition by targeting hedgehog signaling in liver fibrosis. J Gastroenterol Hepatol 2020; 35:482-491. [PMID: 31617598 DOI: 10.1111/jgh.14875] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hepatic fibrosis is the final endpoint for most chronic liver diseases and remains a significant public health problem worldwide. Icariin, a naturally occurring flavonol glucoside, has been reported to exhibit protective effects on liver injury and alleviate liver fibrosis. However, the underlying detail molecular mechanism is not fully revealed. METHODS Mouse primary hepatic stellate cells (HSCs) and carbon tetrachloride (CCL4 )-induced liver fibrosis model in mice were used as in vitro and in vivo models in this study. The expression levels of miR-875-5p were detected by quantitative reverse transcription-PCR. The validation of the direct target of miR-875-5p was through dual-luciferase reporter assay and western blotting assay. The cell proliferation and cell mobility were determined using MTT assay and Transwell migration assay, respectively. RESULTS We found that icariin inhibited epithelial-mesenchymal transition and collagen protein section of HSCs. Icariin exerted hepatoprotective effects on mice model of CCL4 -induced liver fibrosis. Our further results revealed that miR-875-5p was downregulated in human cirrhosis tissues and activated murine HSCs. Icariin induced miR-875-5p upregulation and subsequently decreased glioma-associated oncogene homolog 1 (GLI1) expression through direct binding to the three prime untranslated region of GLI1 mRNA. CONCLUSION Our study highlighted the potential therapeutic application of icariin for liver fibrosis management.
Collapse
Affiliation(s)
- Lei Ye
- First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Zhejiang, China
| | - Yaping Yu
- First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Zhejiang, China
| | - Yanping Zhao
- First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Zhejiang, China
| |
Collapse
|
39
|
Buniatian GH, Weiskirchen R, Weiss TS, Schwinghammer U, Fritz M, Seferyan T, Proksch B, Glaser M, Lourhmati A, Buadze M, Borkham-Kamphorst E, Gaunitz F, Gleiter CH, Lang T, Schaeffeler E, Tremmel R, Cynis H, Frey WH, Gebhardt R, Friedman SL, Mikulits W, Schwab M, Danielyan L. Antifibrotic Effects of Amyloid-Beta and Its Loss in Cirrhotic Liver. Cells 2020; 9:cells9020452. [PMID: 32089540 PMCID: PMC7072823 DOI: 10.3390/cells9020452] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/06/2020] [Accepted: 02/13/2020] [Indexed: 12/15/2022] Open
Abstract
The function and regulation of amyloid-beta (Aβ) in healthy and diseased liver remains unexplored. Because Aβ reduces the integrity of the blood-brain barrier we have examined its potential role in regulating the sinusoidal permeability of normal and cirrhotic liver. Aβ and key proteins that generate (beta-secretase 1 and presenilin-1) and degrade it (neprilysin and myelin basic protein) were decreased in human cirrhotic liver. In culture, activated hepatic stellate cells (HSC) internalized Aβ more efficiently than astrocytes and HSC degraded Aβ leading to suppressed expression of α-smooth muscle actin (α-SMA), collagen 1 and transforming growth factor β (TGFβ). Aβ also upregulated sinusoidal permeability marker endothelial NO synthase (eNOS) and decreased TGFβ in cultured human liver sinusoidal endothelial cells (hLSEC). Liver Aβ levels also correlate with the expression of eNOS in transgenic Alzheimer’s disease mice and in human and rodent cirrhosis/fibrosis. These findings suggest a previously unexplored role of Aβ in the maintenance of liver sinusoidal permeability and in protection against cirrhosis/fibrosis via attenuation of HSC activation.
Collapse
Affiliation(s)
- Gayane Hrachia Buniatian
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
- H. Buniatian Institute of Biochemistry, National Academy of Sciences of the Republic of Armenia (NAS RA), Yerevan 0014, Armenia;
- Correspondence: (G.H.B.); (L.D.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, 52074 Aachen, Germany; (R.W.); (E.B.-K.)
| | - Thomas S. Weiss
- Children’s University Hospital (KUNO), University of Regensburg, 93053 Regensburg, Germany;
| | - Ute Schwinghammer
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
| | - Martin Fritz
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
| | - Torgom Seferyan
- H. Buniatian Institute of Biochemistry, National Academy of Sciences of the Republic of Armenia (NAS RA), Yerevan 0014, Armenia;
| | - Barbara Proksch
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
| | - Michael Glaser
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
| | - Ali Lourhmati
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
| | - Marine Buadze
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
| | - Erawan Borkham-Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, 52074 Aachen, Germany; (R.W.); (E.B.-K.)
| | - Frank Gaunitz
- Department of Neurosurgery, University Hospital of Leipzig, 04103 Leipzig, Germany;
| | - Christoph H. Gleiter
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
| | - Thomas Lang
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany, and University of Tuebingen, 72076 Tuebingen, Germany; (T.L.); (E.S.); (R.T.)
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany, and University of Tuebingen, 72076 Tuebingen, Germany; (T.L.); (E.S.); (R.T.)
| | - Roman Tremmel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany, and University of Tuebingen, 72076 Tuebingen, Germany; (T.L.); (E.S.); (R.T.)
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle, Germany;
| | - William H. Frey
- Center for Memory & Aging, HealthPartners Neuroscience Center, St. Paul, MN 55130, USA;
| | - Rolf Gebhardt
- Rudolf-Schönheimer Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany;
| | - Scott L. Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA;
| | - Wolfgang Mikulits
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria;
| | - Matthias Schwab
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany, and University of Tuebingen, 72076 Tuebingen, Germany; (T.L.); (E.S.); (R.T.)
- Department of Pharmacy and Biochemistry, University of Tuebingen, 72076 Tuebingen, Germany
- Departments of Biochemistry and Clinical Pharmacology, and Neuroscience Laboratory, Yerevan State Medical University, Yerevan 0025, Armenia
| | - Lusine Danielyan
- Department of Clinical Pharmacology, University Hospital of Tübingen, 72076 Tübingen, Germany; (U.S.); (M.F.); (B.P.); (M.G.); (A.L.); (M.B.); (C.H.G.); (M.S.)
- Departments of Biochemistry and Clinical Pharmacology, and Neuroscience Laboratory, Yerevan State Medical University, Yerevan 0025, Armenia
- Correspondence: (G.H.B.); (L.D.)
| |
Collapse
|
40
|
Identification of Ppar γ-modulated miRNA hubs that target the fibrotic tumor microenvironment. Proc Natl Acad Sci U S A 2019; 117:454-463. [PMID: 31871210 PMCID: PMC6955372 DOI: 10.1073/pnas.1909145117] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Liver fibrosis interferes with normal organ function and supports tumor development in the liver. We uncovered a role of miRNAs in controlling liver fibrosis. In a comprehensive and systematic analysis we specify miRNA activities in targeting the fibrotic cellular microenvironment of the liver, in both mice and humans. We reveal and validate a complex network of 8 functionally connected miRNAs and 54 target genes to regulate structural, signaling, and remodeling components of the fibrotic extracellular matrix. We identify expression of this miRNA network to be controlled by the transcription factor Pparγ. Thus, we expand the antifibrotic function of Pparγ to controlling the synthesis of an antifibrotic miRNA network. This network may serve as a therapeutic target in antifibrotic therapies. Liver fibrosis interferes with normal liver function and facilitates hepatocellular carcinoma (HCC) development, representing a major threat to human health. Here, we present a comprehensive perspective of microRNA (miRNA) function on targeting the fibrotic microenvironment. Starting from a murine HCC model, we identify a miRNA network composed of 8 miRNA hubs and 54 target genes. We show that let-7, miR-30, miR-29c, miR-335, and miR-338 (collectively termed antifibrotic microRNAs [AF-miRNAs]) down-regulate key structural, signaling, and remodeling components of the extracellular matrix. During fibrogenic transition, these miRNAs are transcriptionally regulated by the transcription factor Pparγ and thus we identify a role of Pparγ as regulator of a functionally related class of AF-miRNAs. The miRNA network is active in human HCC, breast, and lung carcinomas, as well as in 2 independent mouse liver fibrosis models. Therefore, we identify a miRNA:mRNA network that contributes to formation of fibrosis in tumorous and nontumorous organs of mice and humans.
Collapse
|
41
|
Wu H, Chen G, Wang J, Deng M, Yuan F, Gong J. TIM-4 interference in Kupffer cells against CCL4-induced liver fibrosis by mediating Akt1/Mitophagy signalling pathway. Cell Prolif 2019; 53:e12731. [PMID: 31755616 PMCID: PMC6985653 DOI: 10.1111/cpr.12731] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/13/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES T-cell immunoglobulin domain and mucin domain-4 (TIM-4) is selectively expressed on antigen-presenting cells (APCs) and modulates various immune responses. However, the role of TIM-4 expressed by Kupffer cells (KCs) in liver fibrosis remains unclear. The present study aimed to explore whether and how TIM-4 expressed by KCs is involved in liver fibrosis. MATERIALS AND METHODS Mice chronic liver fibrosis models were established and divided into the olive-induced control group, CCL4-induced control group, olive-induced TIM-4 interference group and CCL4-induced TIM-4 interference group. Different techniques were used to monitor the fibrotic effects of TIM-4, including histopathological assays, Western blotting, ELISA and transmission electron microscopy. Additionally, mice liver transplant models were established to determine the fibrotic effects of TIM-4 on fibrosis after liver transplantation (LT). RESULTS We found that the induction of liver fibrosis by CCL4 was associated with TIM-4 expression in KCs. TIM-4 interference essentially contributed to liver fibrosis resolution. KCs from the TIM-4 interference group had decreased levels of pro-fibrotic markers, reduced TGF-β1 secretion and inhibited hepatic stellate cell (HSC) differentiation into myofibroblast-like cells. In addition, we used GdCl3 to verify that KCs are the primary source of TGF-β1 during fibrosis progression. Moreover, KCs from CCL4-induced mice showed increased ROS production, mitophagy activation and TGF-β1 secretion. However, TIM-4 interference in the KCs inhibited Akt1-mediated ROS production, resulting in the suppression of PINK1, Parkin and LC3-II/I activation and the reduction of TGF-β1 secretion during liver fibrosis. Additionally, TIM-4 interference potentially attenuated development of fibrosis after LT. CONCLUSIONS Our findings revealed the underlying mechanisms of TIM-4 interference in KCs to mitigate liver fibrosis.
Collapse
Affiliation(s)
- Hao Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guoyong Chen
- Department of Hepatobiliary and pancreatic surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jingyuan Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minghua Deng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangchao Yuan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Zhang Z, Guo M, Shen M, Li Y, Tan S, Shao J, Zhang F, Chen A, Wang S, Zheng S. Oroxylin A regulates the turnover of lipid droplet via downregulating adipose triglyceride lipase (ATGL) in hepatic stellate cells. Life Sci 2019; 238:116934. [PMID: 31610205 DOI: 10.1016/j.lfs.2019.116934] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/26/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Proliferation and differentiation of hepatic stellate cells (HSCs) are the most noticeable events in hepatic fibrosis, in which the loss of lipid droplets (LDs) is the most important feature. However, the complex mechanisms of LD disappearance have not been fully elucidated. In the current study, we investigated whether oroxylin A has the pharmacological activity of reversing LDs in activated HSCs, and further examined its potential molecular mechanisms. Using genetic, pharmacological, and molecular biological measure, we found that LD content significantly decreased during HSC activation, whereas oroxylin A markedly reversed LD content in activated HSCs. Interestingly, oroxylin A treatment observably decreased the expression of adipose triglyceride lipase (ATGL) without large differences in classical LD synthesis pathway, LD-related transcription factors, and autophagy pathway. ATGL overexpression could completely impair the effect of oroxylin A on reversing LD content. Importantly, reactive oxygen species (ROS) signaling pathway mediated oroxylin A-induced ATGL downregulation and LD revision in activated HSCs. ROS specific stimulant buthionine sulfoximine (BSO) could dramatically diminish the antioxidant effect of oroxylin A, and in turn, abolish reversal effect of oroxylin A on LD content. Conversely, ROS specific scavenger N-acetyl cystenine (NAC) can significantly enhance the pharmacological effect of oroxylin A on LD revision. Taken together, our study reveals the important molecular mechanism of anti-fibrosis effect of oroxylin A, and also suggests that ROS-ATGL pathway is a potential target for reversing LDs.
Collapse
Affiliation(s)
- Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mei Guo
- Department of Pathogenic biology and Immunology, Medical School, Southeast University, Nanjing, 210009, China
| | - Min Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yujia Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shanzhong Tan
- Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis, MO, 63104, USA
| | - Shijun Wang
- Shandong co-innovation center of TCM formula, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
43
|
Zhai X, Wang W, Dou D, Ma Y, Gang D, Jiang Z, Shi B, Jin B. A novel technique to prepare a single cell suspension of isolated quiescent human hepatic stellate cells. Sci Rep 2019; 9:12757. [PMID: 31485000 PMCID: PMC6726602 DOI: 10.1038/s41598-019-49287-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 08/20/2019] [Indexed: 01/17/2023] Open
Abstract
To explore a simple and easy-to-learn procedure for the isolation of human quiescent hepatic stellate cells (HSCs) that requires no advanced training. Thus reducing costs and increasing efficiency. This protocol will provide sufficient primary cells with minimal contaminants for future basic research on diseases associated with human HSCs. Normal liver tissues were isolated from patients undergoing hepatic hemangioma resection, and a single cell suspension of these tissues was prepared using the Gentle MACS tissue processor. By using this method, the difficulty of the procedure was reduced, fewer cells were lost during the preparation treatments, and the maximal activity of single cells was maintained. Following preparation of the cell suspension, the HSCs were further isolated using a Nycodenz density gradient. Cell viability was examined by trypan blue staining, and the purity of the quiescent human HSCs was determined by autofluorescence and oil red O staining. Activated and quiescent human HSCs were identified using immunofluorescence and Western blotting. The cell cycle distribution in activated and quiescent human HSCs was analyzed by flow cytometry.The recovery rate of the HSCs was approximately (2.1 ± 0.23) × 106 of tissue, with 94.43 ± 1.89% cell viability and 93.8 ± 1.52% purity. The technique used in this study is a simple, high-yield, and repeatable method for HSC isolation that is worthy of recommendation.
Collapse
Affiliation(s)
- Xiangyu Zhai
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Wei Wang
- School of medicine, Shandong University, Jinan, China
| | - Dandan Dou
- School of basic medical sciences, Shandong University, Jinan, China
| | - Yunlong Ma
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Du Gang
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Zhengchen Jiang
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Binyao Shi
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China
| | - Bin Jin
- Department of general surgery, Qilu hospital of Shandong University, Jinan, China.
| |
Collapse
|
44
|
Meurer S, Wimmer AE, Leur EVD, Weiskirchen R. Endoglin Trafficking/Exosomal Targeting in Liver Cells Depends on N-Glycosylation. Cells 2019; 8:cells8090997. [PMID: 31466384 PMCID: PMC6769735 DOI: 10.3390/cells8090997] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023] Open
Abstract
Injury of the liver involves a wound healing partial reaction governed by hepatic stellate cells and portal fibroblasts. Individual members of the transforming growth factor-β (TGF-β) superfamily including TGF-β itself and bone morphogenetic proteins (BMP) exert diverse and partially opposing effects on pro-fibrogenic responses. Signaling by these ligands is mediated through binding to membrane integral receptors type I/type II. Binding and the outcome of signaling is critically modulated by Endoglin (Eng), a type III co-receptor. In order to learn more about trafficking of Eng in liver cells, we investigated the membranal subdomain localization of full-length (FL)-Eng. We could show that FL-Eng is enriched in Caveolin-1-containing sucrose gradient fractions. Since lipid rafts contribute to the pool of exosomes, we could consequently demonstrate for the first time that exosomes isolated from cultured primary hepatic stellate cells and its derivatives contain Eng. Moreover, via adenoviral overexpression, we demonstrate that all liver cells have the capacity to direct Eng to exosomes, irrespectively whether they express endogenous Eng or not. Finally, we demonstrate that block of N-glycosylation does not interfere with dimerization of the receptor, but abrogates the secretion of soluble Eng (sol-Eng) and prevents exosomal targeting of FL-Eng.
Collapse
Affiliation(s)
- Steffen Meurer
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany.
| | - Almut Elisabeth Wimmer
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany
| | - Eddy van de Leur
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany
| | - Ralf Weiskirchen
- RWTH University Hospital Aachen, Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, D-52074 Aachen, Germany.
| |
Collapse
|
45
|
Zhang WJ, Fang ZM, Liu WQ. NLRP3 inflammasome activation from Kupffer cells is involved in liver fibrosis of Schistosoma japonicum-infected mice via NF-κB. Parasit Vectors 2019; 12:29. [PMID: 30635040 PMCID: PMC6330481 DOI: 10.1186/s13071-018-3223-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND NOD-like receptor protein 3 (NLRP3) inflammasome was reported as expressed in schistosomiasis-induced liver fibrosis (SSLF). We used an NLRP3 inflammasome inhibitor, MCC950, to investigate whether it inhibited liver fibrosis, and explored the preliminary molecular mechanism. METHODS BALB/c mice were infected with 15 cercariae through the abdominal skin. They received intraperitoneal injections of MCC950 on the day of infection and at day 22 post-infection. We examined their SSLF phenotype and the effect on liver fibrosis, primary Kupffer cells (KCs), and HSCs. Human hepatic stellate cell lines (human LX-2 cells) were treated with soluble egg antigen (SEA) released from the eggs. We then determined the expression of NLRP3 inflammasome and liver fibrosis-associated markers, liver granuloma and ALT/AST. RESULTS NLRP3 inflammasome expression in the liver was significantly increased, and eosinophilic granuloma and collagen deposition were found around the eggs in mice infected for 56 days. Additionally, IL-1β, ALT/AST in plasma, and NF-κB in liver tissue and in KCs were all greatly significantly increased. The above-mentioned indicators were largely reduced in mice treated with MCC950 on the day of infection. In vitro, lipopolysaccharide (LPS)/SEA could induce LX-2 cells to express NLRP3 and fibrosis markers, and the SEA-treated group was reversed by MCC950. Furthermore, NLRP3 inflammasome and liver fibrosis-associated markers were both increased in the primary KCs and HSCs isolated from infected mice. However, this effect was not observed in the same cells from the mice treated with MCC950 on the day of infection. Contrary to the aforementioned results, MCC950 treatment at day 22 post-infection aggravated this process. Surprisingly, NLRP3 inflammasome was involved in liver fibrosis mostly from KCs. CONCLUSIONS MCC950 acts dually on SSLF pathology and fibrosis in infected mice. Although MCC950 treatment improved SSLF on the day of infection, it exacerbated the pathological effects at day 22 post-infection. These dual effects were mediated via NF-κB. Moreover, NLRP3 inflammasome mainly came from KCs. Our results suggest that blocking NLRP3 on the day of infection may prove to be a promising direction in preventing SSLF.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, Hubei, People's Republic of China
| | - Zheng-Ming Fang
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, Hubei, People's Republic of China
| | - Wen-Qi Liu
- Department of Parasitology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
46
|
Enguita M, Razquin N, Pamplona R, Quiroga J, Prieto J, Fortes P. The cirrhotic liver is depleted of docosahexaenoic acid (DHA), a key modulator of NF-κB and TGFβ pathways in hepatic stellate cells. Cell Death Dis 2019; 10:14. [PMID: 30622239 PMCID: PMC6325107 DOI: 10.1038/s41419-018-1243-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023]
Abstract
Liver cirrhosis results from chronic hepatic damage and is characterized by derangement of the organ architecture with increased liver fibrogenesis and defective hepatocellular function. It frequently evolves into progressive hepatic insufficiency associated with high mortality unless liver transplantation is performed. We have hypothesized that the deficiency of critical nutrients such as essential omega-3 fatty acids might play a role in the progression of liver cirrhosis. Here we evaluated by LC-MS/MS the liver content of omega-3 docosahexaenoic fatty acid (DHA) in cirrhotic patients and investigated the effect of DHA in a murine model of liver injury and in the response of hepatic stellate cells (HSCs) (the main producers of collagen in the liver) to pro-fibrogenic stimuli. We found that cirrhotic livers exhibit a marked depletion of DHA and that this alteration correlates with the progression of the disease. Administration of DHA exerts potent anti-fibrogenic effects in an acute model of liver damage. Studies with HSCs show that DHA inhibits fibrogenesis more intensely than other omega-3 fatty acids. Data from expression arrays revealed that DHA blocks TGFβ and NF-κB pathways. Mechanistically, DHA decreases late, but not early, SMAD3 nuclear accumulation and inhibits p65/RelA-S536 phosphorylation, which is required for HSC survival. Notably, DHA increases ADRP expression, leading to the formation of typical quiescence-associated perinuclear lipid droplets. In conclusion, a marked depletion of DHA is present in the liver of patients with advanced cirrhosis. DHA displays anti-fibrogenic activities on HSCs targeting NF-κB and TGFβ pathways and inducing ADPR expression and quiescence in these cells.
Collapse
Affiliation(s)
- Mónica Enguita
- Department of Gene Therapy and Hepatology, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Nerea Razquin
- Department of Gene Therapy and Hepatology, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida (IRB), Lleida, Spain
| | - Jorge Quiroga
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,Liver Unit, Clínica Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Pamplona, Spain
| | | | - Puri Fortes
- Department of Gene Therapy and Hepatology, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), Pamplona, Spain. .,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
| |
Collapse
|
47
|
TLR5 silencing reduced hyperammonaemia-induced liver injury by inhibiting oxidative stress and inflammation responses via inactivating NF-κB and MAPK signals. Chem Biol Interact 2018; 299:102-110. [PMID: 30508503 DOI: 10.1016/j.cbi.2018.11.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/20/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Liver injury is a serious threat for human health and life. Toll-like receptor 5 (TLR5) has reported to be a vital mediator in flagellin or tetrachloride (CCl4)-induced liver injury. However, the roles and etiology of TLR5 in hyperammonaemia (HA)-induced liver injury are poor defined. METHODS HA rats were generated by intragastric administration using ammonium chloride solution. Liver status was assessed by haematoxylin and eosin (H&E) staining and measuring serum levels of liver injury markers. Immunohistochemistry (IHC) assay was used to visualize protein expression in tissues. Apoptotic index in tissues was determined by TUNEL assay. RT-qPCR assay was employed to test mRNA expression. Oxidative stress responses was assessed by detecting levels of reactive oxygen species (ROS) and related indicators. NF-κB activity was examined by TransAM NF-κB colorimetric kit. RESULTS TLR5 was highly expressed in liver tissues of HA rats. TLR5 knockdown ameliorated HA-induced liver injury by inhibiting liver cell apoptosis. TLR5 depletion inhibited HA-induced pro-inflammatory cytokine expression in liver tissues, but had no effect on the infiltration of T and macrophage cells into liver tissues. TLR5 silencing impaired HA-induced oxidative stress responses in hepatocytes, but not in hepatic stellate cells (HSCs). TLR5 downregulation inhibited HA-induced activation on TLR5/NF-κB and TLR5/MAPK signaling pathways. CONCLUSION TLR5 silencing reduced HA-induced liver injury by inhibiting hepatocyte apoptosis, oxidative stress and inflammation responses via inactivating NF-κB and MAPK signals, deepening our understanding on the molecular mechanism of HA-induced liver injury and providing a potential therapeutic target for alleviating liver injury.
Collapse
|
48
|
Zhang H, Lin Y, Zhen Y, Hu G, Meng X, Li X, Men X. Therapeutic Effect of Glycyrrhizin Arginine Salt on Rat Cholestatic Cirrhosis and its Mechanism. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:1111-1127. [DOI: 10.1142/s0192415x18500581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To investigate the therapeutic effect of glycyrrhizin arginine salt on rat cholestatic cirrhosis, we subjected male Sprague Dawley rats to common bile duct ligation for 14 days and treated them with distilled water (model group), arginine, or a low or high dose of glycyrrhizin arginine salt by gavage. A sham-operated group was used as a control group. Treatment with glycyrrhizin arginine salt substantially improved animal growth rates, reduced the ratio of liver weight to body weight and decreased total bilirubin, aspartate aminotransferase, 8-isoprostane and malondialdehyde compared with the values measured in the model group. The progress of liver fibrosis, as detected by hematoxylin and eosin and Masson’s trichrome staining, was slower in the glycyrrhizin arginine salt groups than in the model group or the arginine group. Reductions of bile salt pool size, hepatic hydroxyproline content and fibrosis score were also seen in the glycyrrhizin arginine salt groups compared with the model group. Furthermore, glycyrrhizin arginine salt significantly reduced the expression of transforming growth factor [Formula: see text]1 (TGF-[Formula: see text]1), [Formula: see text]-smooth muscle actin, tumor necrosis factor-[Formula: see text] and matrix metalloproteinases 2 and 9. Glycyrrhizin arginine salt also inhibited the expression of [Formula: see text]-SMA and matrix metalloproteinases 2 and 9 in response to TGF-[Formula: see text]1 in LX-2 cells and primary rat hepatic stellate cells and mitigated the cytotoxicity induced by rat bile in HepG2 cells and primary rat hepatocytes.
Collapse
Affiliation(s)
- Huan Zhang
- Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063000, P. R. China
| | - Yajun Lin
- The Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P. R. China
| | - Yongzhan Zhen
- Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063000, P. R. China
| | - Gang Hu
- The Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P. R. China
| | - Xu Meng
- Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063000, P. R. China
| | - Xingxin Li
- Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063000, P. R. China
| | - Xiuli Men
- Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063000, P. R. China
| |
Collapse
|
49
|
Zhang H, Ju B, Nie Y, Song B, Xu Y, Gao P. Adenovirus‑mediated knockdown of activin A receptor type 2A attenuates immune‑induced hepatic fibrosis in mice and inhibits interleukin‑17‑induced activation of primary hepatic stellate cells. Int J Mol Med 2018; 42:279-289. [PMID: 29620144 PMCID: PMC5979935 DOI: 10.3892/ijmm.2018.3600] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/08/2018] [Indexed: 01/20/2023] Open
Abstract
Fibrosis induces a progressive loss of liver function, thus leading to organ failure. Activins are secreted proteins that belong to the transforming growth factor (TGF)-β superfamily, which initiate signaling by binding to their two type II receptors: Activin A receptor type 2A (ACVR2A) and activin A receptor type 2B. Previous studies that have explored the mechanisms underlying immune-induced hepatic fibrosis have mainly focused on TGF-β signaling, not activin signaling. To investigate the role of the activin pathway in this disease, adenovirus particles containing short hairpin (sh)RNA targeting ACVR2A mRNA (Ad-ACVR2A shRNA) were administered to mice, which were chronically treated with concanavalin A (Con A). The pathological changes in the liver were evaluated with hematoxylin/eosin staining, Masson trichrome staining and immunohistochemical assay. The results detected an increase in serum activin A and liver ACVR2A in Con A-treated animals. Conversely, liver function was partially restored and fibrotic injury was attenuated when activin signaling was blocked. In addition, the activation of hepatic stellate cells (HSCs) in response to Con A was suppressed by Ad-ACVR2A shRNA, as evidenced by decreased α-smooth muscle actin, and type I and IV collagen expression. Furthermore, primary mouse HSCs (mHSCs) were activated when exposed to interleukin (IL)-17A or IL-17F, which are two major cytokines produced by cluster of differentiation 4+ T helper 17 cells. The levels of activin A, type I and IV collagen were determined with ELISA kits and the expression of fibrotic molecules was determined with western blot analysis. Conversely, blocking activin/ACVR2A impaired the potency of HSCs to produce collagens in response to IL-17s. In addition, C terminus phosphorylation of Smad2 on Ser465 and Ser467, induced by either Con A in the liver or by IL-17s in mHSCs, was partly inhibited when activin A/ACVR2A signaling was suppressed. Collectively, the present study demonstrated an involvement of activated activin A/ACVR2A/Smad2 signaling in immune-induced hepatic fibrosis.
Collapse
Affiliation(s)
- Hongjun Zhang
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Baoling Ju
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Ying Nie
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Baohui Song
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yuanhong Xu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Ping Gao
- Department of Gastroenterology, Mudanjiang Forestry Central Hospital, Mudanjiang, Heilongjiang 157000, P.R. China
| |
Collapse
|