1
|
Xie L, Chen H, Zhang L, Ma Y, Zhou Y, Yang YY, Liu C, Wang YL, Yan YJ, Ding J, Teng X, Yang Q, Liu XP, Wu J. JCAD deficiency attenuates activation of hepatic stellate cells and cholestatic fibrosis. Clin Mol Hepatol 2024; 30:206-224. [PMID: 38190829 PMCID: PMC11016487 DOI: 10.3350/cmh.2023.0506] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND/AIMS Cholestatic liver diseases including primary biliary cholangitis (PBC) are associated with active hepatic fibrogenesis, which ultimately progresses to cirrhosis. Activated hepatic stellate cells (HSCs) are the main fibrogenic effectors in response to cholangiocyte damage. JCAD regulates cell proliferation and malignant transformation in nonalcoholic steatoheaptitis-associated hepatocellular carcinoma (NASH-HCC). However, its participation in cholestatic fibrosis has not been explored yet. METHODS Serial sections of liver tissue of PBC patients were stained with immunofluorescence. Hepatic fibrosis was induced by bile duct ligation (BDL) in wild-type (WT), global JCAD knockout mice (JCAD-KO) and HSC-specific JCAD knockout mice (HSC-JCAD-KO), and evaluated by histopathology and biochemical tests. In situ-activated HSCs isolated from BDL mice were used to determine effects of JCAD on HSC activation. RESULTS In consistence with staining of liver sections from PBC patients, immunofluorescent staining revealed that JCAD expression was identified in smooth muscle α-actin (α-SMA)-positive fibroblast-like cells and was significantly up-regulated in WT mice with BDL. JCAD deficiency remarkably ameliorated BDL-induced hepatic injury and fibrosis, as documented by liver hydroxyproline content, when compared to WT mice with BDL. Histopathologically, collagen deposition was dramatically reduced in both JCAD-KO and HSC-JCAD-KO mice compared to WT mice, as visualized by Trichrome staining and semi-quantitative scores. Moreover, JCAD deprivation significantly attenuated in situ HSC activation and reduced expression of fibrotic genes after BDL. CONCLUSION JCAD deficiency effectively suppressed hepatic fibrosis induced by BDL in mice, and the underlying mechanisms are largely through suppressed Hippo-YAP signaling activity in HSCs.
Collapse
Affiliation(s)
- Li Xie
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Hui Chen
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Li Zhang
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Yue Ma
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Yuan Zhou
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Yong-Yu Yang
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Chang Liu
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Yu-Li Wang
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
| | - Ya-Jun Yan
- Department of Pathology, Shanghai Fifth People’s Hospital, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jia Ding
- Department of Gastroenterology, Jing’an District Central Hospital, Fudan University, Shanghai, China
| | - Xiao Teng
- HistoIndex Pte Ltd, Singapore, Singapore
| | - Qiang Yang
- Hangzhou Choutu Technology Co., Ltd., Hangzhou, China
| | - Xiu-Ping Liu
- Department of Pathology, Shanghai Fifth People’s Hospital, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jian Wu
- Department of Medical Microbiology & Parasitology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai, China
- Department of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
2
|
Liu R, Zhu M, Chen J, Gai J, Huang J, Zhou Y, Wan Y, Tu C. Identification and Characterization of a Novel Nanobody Against Human CTGF to Reveal Its Antifibrotic Effect in an in vitro Model of Liver Fibrosis. Int J Nanomedicine 2023; 18:5407-5422. [PMID: 37753068 PMCID: PMC10519214 DOI: 10.2147/ijn.s428430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Background No agents are currently available for the treatment or reversal of liver fibrosis. Novel antifibrotic therapies for chronic liver diseases are thus urgently needed. Connective tissue growth factor (CTGF) has been shown to contributes profoundly to liver fibrogenesis, which makes CTGF as a promising target for developing antifibrotic agents. Methods In this study, we identified a novel nanobody (Nb) against human CTGF (anti-CTGF Nb) by phage display using an immunized camel, which showed high affinity and specificity in vitro. LX-2 cells, the immortalized human hepatic stellate cells, were induced by transforming growth factor beta1 (TGFβ1) as an in vitro model of liver fibrosis to verify the antifibrotic activity of the anti-CTGF Nb. Results Our data demonstrated that anti-CTGF Nb effectively alleviated TGFβ1-induced LX-2 cell proliferation, activation, and migration, and promoted the apoptosis of activated LX-2 cells in response to TGFβ1. Moreover, the anti-CTGF Nb remarkably reduced the levels of TGFβ1, Smad2, and Smad3 expression in LX-2 stellate cells stimulated by TGFβ1. Conclusion Taken together, we successfully identified a novel Nb against human CTGF, which exhibited antifibrotic effects in vitro by regulating the biological functions of human stellate cells LX-2.
Collapse
Affiliation(s)
- Rong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
| | - Min Zhu
- Shanghai Novamab Biopharmaceuticals Co., Ltd, Shanghai, 201318, People's Republic of China
| | - Jiaojiao Chen
- Department of Gastroenterology, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, 200434, People's Republic of China
| | - Junwei Gai
- Shanghai Novamab Biopharmaceuticals Co., Ltd, Shanghai, 201318, People's Republic of China
| | - Jing Huang
- Shanghai Novamab Biopharmaceuticals Co., Ltd, Shanghai, 201318, People's Republic of China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
| | - Yakun Wan
- Shanghai Novamab Biopharmaceuticals Co., Ltd, Shanghai, 201318, People's Republic of China
| | - Chuantao Tu
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, People's Republic of China
| |
Collapse
|
3
|
Li X, Chen R, Kemper S, Brigstock DR. Production, Exacerbating Effect, and EV-Mediated Transcription of Hepatic CCN2 in NASH: Implications for Diagnosis and Therapy of NASH Fibrosis. Int J Mol Sci 2023; 24:12823. [PMID: 37629004 PMCID: PMC10454308 DOI: 10.3390/ijms241612823] [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: 04/21/2023] [Revised: 08/05/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by steatosis, hepatocyte ballooning, and inflammation and may progress to include increasingly severe fibrosis, which portends more serious disease and is predictive of patient mortality. Diagnostic and therapeutic options for NASH fibrosis are limited, and the underlying fibrogenic pathways are under-explored. Cell communication network factor 2 (CCN2) is a well-characterized pro-fibrotic molecule, but its production in and contribution to NASH fibrosis requires further study. Hepatic CCN2 expression was significantly induced in NASH patients with F3-F4 fibrosis and was positively correlated with hepatic Col1A1, Col1A2, Col3A1, or αSMA expression. When wild-type (WT) or transgenic (TG) Swiss mice expressing enhanced green fluorescent protein (EGFP) under the control of the CCN2 promoter were fed up to 7 weeks with control or choline-deficient, amino-acid-defined diet with high (60%) fat (CDAA-HF), the resulting NASH-like hepatic pathology included a profound increase in CCN2 or EGFP immunoreactivity in activated hepatic stellate cells (HSC) and in fibroblasts and smooth muscle cells of the vasculature, with little or no induction of CCN2 in other liver cell types. In the context of CDAA-HF diet-induced NASH, Balb/c TG mice expressing human CCN2 under the control of the albumin promoter exhibited exacerbated deposition of interstitial hepatic collagen and activated HSC compared to WT mice. In vitro, palmitic acid-treated hepatocytes produced extracellular vesicles (EVs) that induced CCN2, Col1A1, and αSMA in HSC. Hepatic CCN2 may aid the assessment of NASH fibrosis severity and, together with pro-fibrogenic EVs, is a therapeutic target for reducing NASH fibrosis.
Collapse
Affiliation(s)
- Xinlei Li
- Center for Clinical and Translational Research, The Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.C.); (S.K.); (D.R.B.)
| | - Ruju Chen
- Center for Clinical and Translational Research, The Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.C.); (S.K.); (D.R.B.)
| | - Sherri Kemper
- Center for Clinical and Translational Research, The Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.C.); (S.K.); (D.R.B.)
| | - David R. Brigstock
- Center for Clinical and Translational Research, The Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (R.C.); (S.K.); (D.R.B.)
- Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43212, USA
| |
Collapse
|
4
|
Trampuž SR, van Riet S, Nordling Å, Ingelman-Sundberg M. The Role of CTGF in Liver Fibrosis Induced in 3D Human Liver Spheroids. Cells 2023; 12:cells12020302. [PMID: 36672237 PMCID: PMC9857203 DOI: 10.3390/cells12020302] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Connective tissue growth factor (CTGF) is involved in the regulation of extracellular matrix (ECM) production. Elevated levels of CTGF can be found in plasma from patients with liver fibrosis and in experimental animal models of liver fibrosis, but the exact role of CTGF in, e.g., diet-induced human liver fibrosis is not entirely known. To address this question, we utilized a 3D human liver co-culture spheroid model composed of hepatocytes and non-parenchymal cells, in which fibrosis is induced by TGF-β1, CTGF or free fatty acids (FFA). Treatment of the spheroids with TGF-β1 or FFA increased COL1A1 deposition as well as the expression of TGF-β1 and CTGF. Recombinant CTGF, as well as angiotensin II, caused increased expression and/or production of CTGF, TGF-β1, COL1A1, LOX, and IL-6. In addition, silencing of CTGF reduced both TGF-β1- and FFA-induced COL1A1 deposition. Furthermore, we found that IL-6 induced CTGF, COL1A1 and TGF-β1 production, suggesting that IL-6 is a mediator in the pathway of CTGF-induced fibrosis. Taken together, our data indicate a specific role for CTGF and CTGF downstream signaling pathways for the development of liver inflammation and fibrosis in the human 3D liver spheroid model.
Collapse
Affiliation(s)
- Sara Redenšek Trampuž
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Sander van Riet
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Åsa Nordling
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Magnus Ingelman-Sundberg
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
5
|
Thompson MD, Hinrichs H, Faerber A, Tarr PI, Davidson NO. Maternal obesogenic diet enhances cholestatic liver disease in offspring. J Lipid Res 2022; 63:100205. [PMID: 35341737 PMCID: PMC9046959 DOI: 10.1016/j.jlr.2022.100205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/08/2022] [Accepted: 03/19/2022] [Indexed: 10/25/2022] Open
Abstract
Human and animal model data show that maternal obesity promotes nonalcoholic fatty liver disease in offspring and alters bile acid (BA) homeostasis. Here we investigated whether offspring exposed to maternal obesogenic diets exhibited greater cholestatic injury. We fed female C57Bl6 mice conventional chow (CON) or high fat/high sucrose (HF/HS) diet and then bred them with lean males. Offspring were fed 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) for 2 weeks to induce cholestasis, and a subgroup was then fed CON for an additional 10 days. Additionally, to evaluate the role of the gut microbiome, we fed antibiotic-treated mice cecal contents from CON or HF/HS offspring, followed by DDC for 2 weeks. We found that HF/HS offspring fed DDC exhibited increased fine branching of the bile duct (ductular reaction) and fibrosis but did not differ in BA pool size or intrahepatic BA profile compared to offspring of mice fed CON. We also found that after 10 days recovery, HF/HS offspring exhibited sustained ductular reaction and periportal fibrosis, while lesions in CON offspring were resolved. In addition, cecal microbiome transplant from HF/HS offspring donors worsened ductular reaction, inflammation, and fibrosis in mice fed DDC. Finally, transfer of the microbiome from HF/HS offspring replicated the cholestatic liver injury phenotype. Taken together, we conclude that maternal HF/HS diet predisposes offspring to increased cholestatic injury after DDC feeding and delays recovery after returning to CON diets. These findings highlight the impact of maternal obesogenic diet on hepatobiliary injury and repair pathways during experimental cholestasis.
Collapse
Affiliation(s)
- Michael D Thompson
- Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
| | - Holly Hinrichs
- Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Austin Faerber
- Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Phillip I Tarr
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
Pantoprazole ameliorates liver fibrosis and suppresses hepatic stellate cell activation in bile duct ligation rats by promoting YAP degradation. Acta Pharmacol Sin 2021; 42:1808-1820. [PMID: 34465912 PMCID: PMC8563954 DOI: 10.1038/s41401-021-00754-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is one of the most severe pathologic consequences of chronic liver diseases, and effective therapeutic strategies are urgently needed. Proton pump inhibitors (PPIs) are H+/K+-ATPase inhibitors and currently used to treat acid-related diseases such as gastric ulcers, which have shown other therapeutic effects in addition to inhibiting acid secretion. However, few studies have focused on PPIs from the perspective of inhibiting hepatic fibrosis. In the present study, we investigated the effects of pantoprazole (PPZ), a PPI, against liver fibrosis in a bile duct ligation (BDL) rat model, human hepatic stellate cell (HSC) line LX-2 and mouse primary HSCs (pHSCs), and explored the potential mechanisms underlying the effects of PPZ in vitro and in vivo. In BDL rats, administration of PPZ (150 mg· kg-1· d-1, i.p. for 14 d) significantly attenuated liver histopathological injury, collagen accumulation, and inflammatory responses, and suppressed fibrogenesis-associated gene expression including Col1a1, Acta2, Tgfβ1, and Mmp-2. In LX-2 cells and mouse pHSCs, PPZ (100-300 μM) dose-dependently suppressed the levels of fibrogenic markers. We conducted transcriptome analysis and subsequent validation in PPZ-treated LX-2 cells, and revealed that PPZ inhibited the expression of Yes-associated protein (YAP) and its downstream targets such as CTGF, ID1, survivin, CYR61, and GLI2. Using YAP overexpression and silencing, we demonstrated that PPZ downregulated hepatic fibrogenic gene expression via YAP. Furthermore, we showed that PPZ promoted the proteasome-dependent degradation and ubiquitination of YAP, thus inhibiting HSC activation. Additionally, we showed that PPZ destabilized YAP by disrupting the interaction between a deubiquitinating enzyme OTUB2 and YAP, and subsequently blocked the progression of hepatic fibrosis.
Collapse
|
7
|
Salas A, Vázquez P, Bello AR, Báez D, Almeida TA. Dual agonist-antagonist effect of ulipristal acetate in human endometrium and myometrium. Expert Rev Mol Diagn 2021; 21:851-857. [PMID: 34110938 DOI: 10.1080/14737159.2021.1941878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The aim of this study was to assess the molecular effect of ulipristal acetate (UPA) on gene expression in myometrium and endometrium of patients with symptomatic fibroids. Tissues isolated from four women treated preoperatively with UPA (5 mg) were compared to those from untreated controls using NanoString platform to assess the expression of 75 candidate genes modulated by UPA and ovarian steroids. Deregulated genes were then validated by real-time PCR. In myometrium, UPA exerted an antagonistic effect similar to that observed in fibroids. In UPA-treated endometrium, six genes were identified as highly and significantly upregulated, including matricellular genes CCN1 (54-fold, P = 0.0018) and CCN2 (11-fold, P = 0.00044), Krüppel-like factor 4 (>3-fold, P = 0.0036), and mast cell markers including tryptases TPSAB1/TPSB2 (31-fold, P = 0.023) and carboxypeptidase A (CPA3, 17-fold, P = 0.05). In endometrium, UPA induced the expression of genes involved in fibrogenesis and mast cell function-some of them being widely involved in hepatic injury, which could explain the marked fibrosis and inflammatory cell infiltration observed in explanted livers from patients under UPA treatment.
Collapse
Affiliation(s)
- Ana Salas
- Departamento de Bioquímica, Microbiología, Biología Celular Y Genética, Universidad de La Laguna. Facultad De Ciencias. Sección de Biología, Tenerife, Spain.,Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC). Avda. Astrofísico Fco. Sánchez S/n. 38200. San Cristóbal de La Laguna, Tenerife, Spain
| | - Paula Vázquez
- Departamento de Bioquímica, Microbiología, Biología Celular Y Genética, Universidad de La Laguna. Facultad De Ciencias. Sección de Biología, Tenerife, Spain
| | - Aixa R Bello
- Departamento de Bioquímica, Microbiología, Biología Celular Y Genética, Universidad de La Laguna. Facultad De Ciencias. Sección de Biología, Tenerife, Spain.,Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC). Avda. Astrofísico Fco. Sánchez S/n. 38200. San Cristóbal de La Laguna, Tenerife, Spain
| | - Delia Báez
- Departamento de Obstetricia y Ginecología, Facultad de Ciencias de La Salud, Universidad de La Laguna, Campus de Ofra S/n, Tenerife, Spain
| | - Teresa A Almeida
- Departamento de Bioquímica, Microbiología, Biología Celular Y Genética, Universidad de La Laguna. Facultad De Ciencias. Sección de Biología, Tenerife, Spain.,Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC). Avda. Astrofísico Fco. Sánchez S/n. 38200. San Cristóbal de La Laguna, Tenerife, Spain
| |
Collapse
|
8
|
Yao J, Lin C, Jiang J, Zhang X, Li F, Liu T, Diao H. lncRNA-HEIM Facilitated Liver Fibrosis by Up-Regulating TGF- β Expression in Long-Term Outcome of Chronic Hepatitis B. Front Immunol 2021; 12:666370. [PMID: 34168644 PMCID: PMC8217658 DOI: 10.3389/fimmu.2021.666370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022] Open
Abstract
Background Chronic liver fibrosis is an inevitable stage for the development of patients with chronic hepatitis B (CHB). However, anti-fibrotic therapies have been unsuccessful so far. The biological functions and molecular mechanisms of long non-coding RNAs (lncRNAs) in the host immune system during chronic hepatitis B virus (HBV) infection, especially in fibrosis, are still largely unknown. Method The total RNA of peripheral blood mononuclear cells (PBMCs) from asymptomatic carriers (ASCs) or CHB receiving at least 8 years of anti-viral treatments was analyzed using Arraystar microarray and validated via quantitative real-time PCR (qRT-PCR). Correlation analysis was conducted based on correlation coefficients, Clusterprofile, and RNA Interactome Database (RAID). The functions of lncRNA in monocytes were determined via loss-of-function RNAi or gain-of-function lentivirus assays. The expression levels of mRNAs or proteins were evaluated using qRT-PCR, western blotting assay, or enzyme linked immunosorbent assays (ELISA). Results A total of 1,042 mRNA transcripts (630 up-regulated and 412 down-regulated) were identified being differentially expressed between ASC and CHB patients. Through enrichment analysis we focused on the transforming growth factor beta (TGF-β) signaling pathway and validated their expression in a larger cohort. Moreover, we found that lncRNA ENST00000519726 (lncRNA-HEIM) was highly expressed in monocytes and further up-regulated upon HBV infection. LncRNA-HEIM played an important role in CHB patients with long-term antiviral treatments, and its elevated expression was remarkably correlated with the TGF-β signaling pathway, especially with the two members namely TGF-β and SMAD4. Furthermore, altering the endogenous lncRNA-HEIM level in monocytes significantly affected the production of TGF-β, as well as the fibrosis of hepatic stellate cells by affecting the expression of collagen I and α-smooth muscle actin (α-SMA). Conclusion These findings not only added knowledge to the understanding of the roles of which lncRNA-HEIM played in the activation of HSCs in CHB patients with long-term medication, but also provided a promising therapeutic target in the future treatment for liver fibrosis.
Collapse
Affiliation(s)
- Jian Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chenhong Lin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xujun Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Fengxia Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Tianxing Liu
- Department of Biological Sciences, University of Toronto, Toronto, ON, Canada
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Jensen VS, Fledelius C, Zachodnik C, Damgaard J, Nygaard H, Tornqvist KS, Kirk RK, Viuff BM, Wulff EM, Lykkesfeldt J, Hvid H. Insulin treatment improves liver histopathology and decreases expression of inflammatory and fibrogenic genes in a hyperglycemic, dyslipidemic hamster model of NAFLD. J Transl Med 2021; 19:80. [PMID: 33596938 PMCID: PMC7890970 DOI: 10.1186/s12967-021-02729-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/29/2021] [Indexed: 11/24/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are highly prevalent comorbidities in patients with Type 2 diabetes. While many of these patients eventually will need treatment with insulin, little is known about the effects of insulin treatment on histopathological parameters and hepatic gene expression in diabetic patients with co-existing NAFLD and NASH. To investigate this further, we evaluated the effects of insulin treatment in NASH diet-fed hamsters with streptozotocin (STZ) -induced hyperglycemia. Methods Forty male Syrian hamsters were randomized into four groups (n = 10/group) receiving either a NASH-inducing (high fat, fructose and cholesterol) or control diet (CTRL) for four weeks, after which they were treated with STZ or sham-injected and from week five treated with either vehicle (CTRL, NASH, NASH-STZ) or human insulin (NASH-STZ-HI) for four weeks by continuous s.c. infusion via osmotic minipumps. Results NASH-STZ hamsters displayed pronounced hyperglycemia, dyslipidemia and more severe liver pathology compared to both CTRL and NASH groups. Insulin treatment attenuated dyslipidemia in NASH-STZ-HI hamsters and liver pathology was considerably improved compared to the NASH-STZ group, with prevention/reversal of hepatic steatosis, hepatic inflammation and stellate cell activation. In addition, expression of inflammatory and fibrotic genes was decreased compared to the NASH-STZ group. Conclusions These results suggest that hyperglycemia is important for development of inflammation and profibrotic processes in the liver, and that insulin administration has beneficial effects on liver pathology and expression of genes related to inflammation and fibrosis in a hyperglycemic, dyslipidemic hamster model of NAFLD.
Collapse
Affiliation(s)
- Victoria Svop Jensen
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg, Denmark. .,Diabetes Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Måløv, Denmark.
| | - Christian Fledelius
- Diabetes Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Måløv, Denmark
| | - Christina Zachodnik
- Diabetes Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Måløv, Denmark
| | - Jesper Damgaard
- Diabetes Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Måløv, Denmark
| | - Helle Nygaard
- Diabetes Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Måløv, Denmark
| | | | - Rikke Kaae Kirk
- Pathology & Imaging, Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Måløv, Denmark
| | | | - Erik Max Wulff
- Gubra ApS, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | - Jens Lykkesfeldt
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg, Denmark
| | - Henning Hvid
- Pathology & Imaging, Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Måløv, Denmark
| |
Collapse
|
10
|
Yang DJ, Shi JH, Xia ZP, Guo WZ, Ahmed MS, Zhang SJ. Hepatic connective tissue growth factor expression and regulation differ between non-steatotic and non-alcoholic steatotic livers from brain-dead donor. Sci Rep 2021; 11:3857. [PMID: 33594198 PMCID: PMC7886893 DOI: 10.1038/s41598-021-83516-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/04/2021] [Indexed: 11/09/2022] Open
Abstract
Accurate evaluation of liver steatosis is required from brain-dead donors (BDDs) with nonalcoholic fatty liver disease (NAFLD). Our purposes were to investigate expression and regulation of connective tissue growth factor (CTGF) expression in livers from human and rat after brain death, and further evaluate its potential application. NAFLD and brain death models were established in rats. LX2 cells were cultured under hypoxia/reoxygenation. CTGF protein and mRNA levels were measured in liver samples from BDDs of human and rat by immunohistochemistry and reverse transcription-quantitative polymerase chain reaction. YAP-regulated CTGF expression was investigated in LX2 cells via YAP small interfering RNA and Verteporfin treatment. Blood CTGF level from BDDs was measured by enzyme-linked immunosorbent assay. After brain death, CTGF, transforming growth factor-β and YAP were overexpressed in non-alcoholic steatotic liver, whereas CTGF was downregulated in non-steatotic liver. Time-series analysis revealed that CTGF and YAP expression was comparable, as confirmed by inhibited YAP expression in LX2 cells. CTGF level and NAFLD activity were linearly correlated. CTGF expression and regulation differ between non-steatosis and nonalcoholic steatosis livers from BDDs. CTGF may be an important factor to evaluate graft quality from BDDs with NAFLD.
Collapse
Affiliation(s)
- Dong-Jing Yang
- Department of Hepatobiliary and Pancreatic Surgery, Henan Key Laboratory of Digestive Organ Transplantation and Zhengzhou Key Laboratory for HPB Diseases and Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Ji-Hua Shi
- Department of Hepatobiliary and Pancreatic Surgery, Henan Key Laboratory of Digestive Organ Transplantation and Zhengzhou Key Laboratory for HPB Diseases and Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zong-Ping Xia
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Wen-Zhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, Henan Key Laboratory of Digestive Organ Transplantation and Zhengzhou Key Laboratory for HPB Diseases and Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mohammed Shakil Ahmed
- Institute for Surgical Research, Department of Cardiology and Center for Heart Failure Research, Oslo University Hospital-Rikshospitalet and University of Oslo, Oslo, Norway
| | - Shui-Jun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Henan Key Laboratory of Digestive Organ Transplantation and Zhengzhou Key Laboratory for HPB Diseases and Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
11
|
Xiang D, Zou J, Zhu X, Chen X, Luo J, Kong L, Zhang H. Physalin D attenuates hepatic stellate cell activation and liver fibrosis by blocking TGF-β/Smad and YAP signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153294. [PMID: 32771890 DOI: 10.1016/j.phymed.2020.153294] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/14/2020] [Accepted: 07/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Hepatic fibrosis is considered integral to the progression of chronic liver diseases, as it leads to the development of cirrhosis and hepatocellular carcinoma. The activation of hepatic stellate cells (HSCs) is the dominant event in hepatic fibrogenesis. The transforming growth factor-β1 (TGF-β1) and Yes-associated protein (YAP) pathways play a pivotal role in HSC activation, hepatic fibrosis and cirrhosis progression. Therefore, targeting the TGF-β/Smad and YAP signaling pathways is a promising strategy for antifibrotic therapy. PURPOSE The present study investigated the protective effects of Physalin D (PD), a withanolide isolated from Physalis species (Solanaceae), against liver fibrosis and further elucidated the mechanisms involved in vitro and in vivo. STUDY DESIGN/METHODS We conducted a series of experiments using carbon tetrachloride (CCl4)- and bile duct ligation (BDL)-induced fibrotic mice and cultured LX-2 cells. Serum markers of liver injury, and the morphology, histology and fibrosis of liver tissue were investigated. Western blot assays and quantitative real-time PCR were used to investigate the mechanisms underlying the antifibrotic effects of PD. RESULT PD decreased TGF-β1-induced COL1A1 promoter activity. PD inhibited TGF-β1-induced expression of Collagen I and α-smooth muscle actin (α-SMA) in human hepatic stellate LX-2 cells. PD significantly ameliorated hepatic injury, including transaminase activities, histology, collagen deposition and α-SMA, in CCl4- or BDL-induced mice. Moreover, PD markedly decreased the expression of phosphorylated Smad2/3 in vitro and in vivo. Furthermore, PD significantly decreased YAP protein levels, and YAP knockdown did not further enhance the effects of PD, namely α-SMA inhibition, Collagen I expression and YAP target gene expression in LX-2 cells. CONCLUSION These results clearly show that PD ameliorated experimental liver fibrosis by inhibiting the TGF-β/Smad and YAP signaling pathways, indicating that PD has the potential to effectively treat liver fibrosis.
Collapse
Affiliation(s)
- Dejuan Xiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Zou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoyun Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xinling Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jianguang Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
12
|
Chymase as a Possible Therapeutic Target for Amelioration of Non-Alcoholic Steatohepatitis. Int J Mol Sci 2020; 21:ijms21207543. [PMID: 33066113 PMCID: PMC7589185 DOI: 10.3390/ijms21207543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
The development and progression of non-alcoholic steatohepatitis (NASH) are linked to oxidative stress, inflammation, and fibrosis of the liver. Chymase, a chymotrypsin-like enzyme produced in mast cells, has various enzymatic actions. These actions include activation of angiotensin II, matrix metalloproteinase (MMP)-9, and transforming growth factor (TGF)-β, which are associated with oxidative stress, inflammation, and fibrosis, respectively. Augmentation of chymase activity in the liver has been reported in various NASH models. Generation of hepatic angiotensin II and related oxidative stress is upregulated in NASH but attenuated by treatment with a chymase inhibitor. Additionally, increases in MMP-9 and accumulation of inflammatory cells are observed in NASH but are decreased by chymase inhibitor administration. TGF-β and collagen I upregulation in NASH is also attenuated by chymase inhibition. These results in experimental NASH models demonstrate that a chymase inhibitor can effectively ameliorate NASH via the reduction of oxidative stress, inflammation, and fibrosis. Thus, chymase may be a therapeutic target for amelioration of NASH.
Collapse
|
13
|
Chiabotto G, Camussi G, Bruno S. Role of ncRNAs in modulation of liver fibrosis by extracellular vesicles. ACTA ACUST UNITED AC 2020. [DOI: 10.1186/s41544-020-00050-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
AbstractExtracellular vesicles (EVs) are small membrane vesicles carrying bioactive lipids, proteins and nucleic acids of the cell of origin. In particular, EVs carry non-coding RNAs (ncRNAs) and the vesicle membrane may protect them from degradation. Once released within the extracellular space, EVs can transfer their cargo, including ncRNAs, to neighboring or distant cells, thus inducing phenotypical and functional changes that may be relevant in several physio-pathological conditions. This review provides an overview of the role of EV-carried ncRNAs in the modulation of liver fibrosis. In particular, we focused on EV-associated microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) involved into the development of liver fibrosis and on the potential use of EV-associated ncRNAs as diagnostic and prognostic biomarkers of liver fibrosis.
Collapse
|
14
|
Dessein H, Duflot N, Romano A, Opio C, Pereira V, Mola C, Kabaterene N, Coutinho A, Dessein A. Genetic algorithms identify individuals with high risk of severe liver disease caused by schistosomes. Hum Genet 2020; 139:821-831. [PMID: 32277285 DOI: 10.1007/s00439-020-02160-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/28/2020] [Indexed: 02/06/2023]
Abstract
Schistosomes induce severe hepatic disease, which is fatal in 2-10% of cases, mortality being higher in cases of co-infection with HBV or HCV. Hepatic disease occurs as a consequence of the chronic inflammation caused by schistosome eggs trapped in liver sinusoids. In certain individuals, the repair process leads to a massive accumulation of fibrosis in the periportal spaces. We and others have shown that genetic variants play a crucial role in disease progression from mild to severe fibrosis and explain why hepatic fibrosis progresses rapidly in certain subjects only. We will review here published findings concerning the strategies that have been used in the analysis of hepatic fibrosis in schistosome-infected individuals, the genetic variants that have associated with fibrosis, and variants in new pathways crucial for fibrosis progression. Together, these studies show that the development of fibrosis is under the tight genetic control of various common variants with moderate effects. This polygenic control has made it possible to develop models that identify schistosome-infected individual at risk of severe hepatic disease. We discuss the performances and limitations of these models.
Collapse
Affiliation(s)
- Hélia Dessein
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France
| | - Nicolas Duflot
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France
| | - Audrey Romano
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France
| | - Christopher Opio
- Department of Medicine, Mulago Hospital, Makerere University College of Health Sciences, Kampala, Uganda
| | - Valeria Pereira
- Instituto Aggeu Magalhães, Fiocruz, Fundaçao Oswaldo Cruz, Av. Professor Moraes Rego, S/N Cidade Universitária, Recife, PE, 50740-465, Brazil
| | - Carla Mola
- Instituto Aggeu Magalhães, Fiocruz, Fundaçao Oswaldo Cruz, Av. Professor Moraes Rego, S/N Cidade Universitária, Recife, PE, 50740-465, Brazil
| | - Narcis Kabaterene
- Vector Control Division Uganda, Ministry of Health, Queen's Ln, Kampala, Uganda
| | - Ana Coutinho
- Fundação Oswaldo Cruz Rio de Janeiro, Av. Brasil, 4365, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Alain Dessein
- BILHI Genetics, 60 Avenue André Roussin, 13016, Marseille, France.
- UMR_S906-Génétique Et Immunologie Des Maladies Parasitaires, Aix Marseille Université-INSERM, Marseille, France.
| |
Collapse
|
15
|
Chen L, Brenner DA, Kisseleva T. Combatting Fibrosis: Exosome-Based Therapies in the Regression of Liver Fibrosis. Hepatol Commun 2019; 3:180-192. [PMID: 30766956 PMCID: PMC6357832 DOI: 10.1002/hep4.1290] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/24/2018] [Indexed: 12/14/2022] Open
Abstract
Hepatic fibrosis results from chronic injury and inflammation in the liver and leads to cirrhosis, liver failure, and portal hypertension. Understanding the molecular mechanisms underlying hepatic fibrosis has advanced the prospect of developing therapies for regression of the disease. Resolution of fibrosis requires a reduction of proinflammatory and fibrogenic cytokines, a decrease in extracellular matrix (ECM) protein production, an increase in collagenase activity, and finally, a disappearance of activated myofibroblasts. Exosomes are nanovesicles of endocytic origin secreted by most cell types. They epigenetically reprogram and alter the phenotype of their recipient cells and hold great promise for the reversal of fibrosis. Recent studies have shown that exosomes function as conduits for intercellular transfer and contain all the necessary components to induce resolution of fibrosis, including the ability to (1) inhibit macrophage activation and cytokine secretion, (2) remodel ECM production and decrease fibrous scars, and (3) inactivate hepatic stellate cells, a major myofibroblast population. Here, we discuss the research involving the regression of hepatic fibrosis. We focus on the newly discovered roles of exosomes during fibrogenesis and as a therapy for fibrosis reversal. We also emphasize the novel discoveries of exosome-based antifibrotic treatments in vitro and in vivo.
Collapse
Affiliation(s)
- Li Chen
- Department of MedicineUniversity of California San DiegoLa JollaCA
| | - David A. Brenner
- Department of MedicineUniversity of California San DiegoLa JollaCA
| | | |
Collapse
|
16
|
Takagi T, Suzuki R, Sugimoto M, Konno N, Sato Y, Irie H, Watanabe K, Nakamura J, Takasumi M, Hikichi T, Ohira H. Non‑specific elevation of serum Mac‑2 binding protein glycosylation isomer levels in patients with biliary disease. Mol Clin Oncol 2018; 10:168-172. [DOI: 10.3892/mco.2018.1750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/10/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Tadayuki Takagi
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Rei Suzuki
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Mitsuru Sugimoto
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Naoki Konno
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Yuki Sato
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Hiroki Irie
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Ko Watanabe
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Jun Nakamura
- Department of Endoscopy, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Mika Takasumi
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Takuto Hikichi
- Department of Endoscopy, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| |
Collapse
|
17
|
Xu Z, Li T, Li M, Yang L, Xiao R, Liu L, Chi X, Liu D. eRF3b-37 inhibits the TGF-β1-induced activation of hepatic stellate cells by regulating cell proliferation, G0/G1 arrest, apoptosis and migration. Int J Mol Med 2018; 42:3602-3612. [PMID: 30272252 DOI: 10.3892/ijmm.2018.3900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/20/2018] [Indexed: 11/05/2022] Open
Abstract
The therapeutic management of liver fibrosis remains an unresolved clinical problem. The activation of hepatic stellate cells (HSCs) serves a pivotal role in the formation of liver fibrosis. In our previous study, matrix‑assisted laser desorption/ionization time‑of‑flight mass spectrometry (MALDI‑TOF MS) was employed to identify potential serum markers for liver cirrhosis, such as eukaryotic peptide chain releasing factor 3b polypeptide (eRF3b‑37), which was initially confirmed by our group to serve a protective role in liver tissues in a C‑C motif chemokine ligand 4‑induced liver cirrhosis mouse model. Therefore, eRF3b‑37 was hypothesized to affect the activation state of HSCs, which was determined by the expression of pro‑fibrogenic associated factors in HSCs. In the present study, peptide synthesis technology was employed to elucidate the role of eRF3b‑37 in the expression of pro‑fibrogenic factors induced by transforming growth factor‑β1 (TGF‑β1) in LX‑2 cells that were treated with either control, TGF‑β1 and TGF‑β1+eRF3b‑37. 3‑(4,5‑Dimethyl‑2‑thiazolyl)‑2,5‑diphenyltetrazolium bromide and flow cytometric assays, and fluorescent microscope examinations were performed to evaluate the effects of eRF3b‑37 on proliferation viability, G0/G1 arrest, apoptosis and cell migration. The results of the present study indicated that eRF3b‑37 inhibited the activation of HSCs. The increased mRNA and protein expression of the pro‑fibrogenic factors collagen I, connective tissue growth factor and α‑smooth muscle actin (SMA) stimulated by TGF‑β1 were reduced by eRF3b‑37 via the following mechanisms: i) Inhibiting LX‑2 cell proliferation, leading to G0/G1 cell cycle arrest and inhibition of DNA synthesis by downregulating the mRNA expressions of Cyclin D1 and cyclin dependent kinase‑4, and upregulating the levels of P21; ii) increasing cell apoptosis by upregulating the mRNA level of B‑cell lymphoma-2 (Bcl‑2)‑associated X protein (Bax) and Fas, and downregulating the expression of Bcl‑2; and iii) reducing cell migration by downregulating the mRNA and protein expression of α‑SMA. In addition, eRF3b‑37 is thought to serve a role in HSCs by inhibiting TGF‑β signaling. Therefore, eRF3b‑37 may be a novel therapeutic agent for targeting HSCs for hepatic fibrosis.
Collapse
Affiliation(s)
- Zhengrong Xu
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Tao Li
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Man Li
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lei Yang
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Rudan Xiao
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Li Liu
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Xin Chi
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Dianwu Liu
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
18
|
Udomsinprasert W, Vejchapipat P, Klaikeaw N, Chongsrisawat V, Poovorawan Y, Honsawek S. Hepatic autotaxin overexpression in infants with biliary atresia. PeerJ 2018; 6:e5224. [PMID: 30065861 PMCID: PMC6063256 DOI: 10.7717/peerj.5224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022] Open
Abstract
Background Autotaxin (ATX) is a secreted glycoprotein that is involved in the development of hepatic fibrogenesis via the enzymatic production of lysophosphatidic acid. The aim of this study was to investigate hepatic expression of ATX in biliary atresia (BA) compared with non-BA liver controls and to examine the association between ATX expression and clinical outcome in BA. Methods Liver specimens from BA infants (n = 20) were compared with samples from infants who underwent liver biopsy for reasons other than BA (n = 14) and served as controls. Relative mRNA and protein expression of ATX were quantified using real-time polymerase chain reaction (PCR) and immunohistochemistry. Masson’s Trichrome staining was performed to determine the degree of liver fibrosis. Results Quantitative real-time PCR demonstrated overexpression of ATX mRNA in BA livers. In immunohistochemical evaluation, ATX was positively stained on the hepatic parenchyma and the biliary epithelium in BA patients, as compared to non-BA controls. The immunostaining score of ATX in BA livers was also significantly higher than that observed in non-BA livers (P < 0.001). Subgroup analysis revealed that ATX expression in the patients with poor outcomes was significantly greater than in those with good outcomes (P = 0.03). Additionally, there was a positive correlation between hepatic ATX expression and Metavir fibrosis stage in BA livers (r = 0.79, P < 0.001). Discussion This study found that mRNA and protein expression of ATX were increased in BA livers. High hepatic ATX expression at the time of Kasai operation was associated with liver fibrosis and outcome in BA, suggesting that ATX may serve a role as a promising biomarker of the prognosis in biliary atresia.
Collapse
Affiliation(s)
- Wanvisa Udomsinprasert
- Osteoarthritis and Musculoskeleton Research Unit, Department of Biochemistry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, Thailand.,Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Paisarn Vejchapipat
- Department of Surgery, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, Thailand
| | - Naruemon Klaikeaw
- Department of Pathology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, Thailand
| | - Voranush Chongsrisawat
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Sittisak Honsawek
- Osteoarthritis and Musculoskeleton Research Unit, Department of Biochemistry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
19
|
Abstract
Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.
Collapse
Affiliation(s)
- Mara H Sherman
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon 97201, USA;
| |
Collapse
|
20
|
Offor O, Utay N, Reynoso D, Somasunderam A, Currier J, Lake J. Adiponectin and the steatosis marker Chi3L1 decrease following switch to raltegravir compared to continued PI/NNRTI-based antiretroviral therapy. PLoS One 2018; 13:e0196395. [PMID: 29746485 PMCID: PMC5944924 DOI: 10.1371/journal.pone.0196395] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/12/2018] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND People with HIV are at for metabolic syndrome (MetS) and fatty liver disease, but the role of Antiretroviral therapy (ART) is poorly understood. MetS and fatty liver disease been associated with changes in adiponectin, soluble ST2 (sST2), chitinase 3-like 1 (Chi3L1), hyaluronic acid (HA), tissue inhibitor of metalloproteinase-1 (TIMP-1), lysyl oxidase-like-2 (LOXL2) and transforming growth factor β (TGF-β) concentrations in HIV-uninfected populations. Protease (PI) and non-nucleoside reverse transcriptase inhibitors (NNRTI) may contribute to these comorbidities, but the effects of switching from PI- or NNRTI to raltegravir (RAL) on these biomarkers is unknown. METHODS Cryopreserved plasma was obtained from a completed, prospective trial of HIV-infected women with central adiposity on NNRTI- or PI-based ART during which they were randomized to remain on their current ART or switch to a RAL based regimen. Biomarker concentrations were quantified using ELISA and Multiplex assays at baseline and 24 weeks after randomization. Wilcoxon-signed rank test evaluated within-group changes, Spearman and linear regression models evaluated correlations between biomarkers and clinical covariates. RESULTS Participants had a median age of 43 years, CD4+ T lymphocyte count 558 cells/mm3 and BMI 32 kg/m2; 35% met criteria for MetS. At baseline, higher adiponectin levels correlated with higher Chi3L1 levels (r = 0.42, p = 0.02), as did declines after 24 weeks (r = 0.40, p = 0.03). Changes in sST2 correlated with changes in Chi3L1 (r = 0.43, p = 0.02) and adiponectin (r = 0.40, p = 0.03). Adiponectin and Chi3L1 levels decreased significantly in women switched to RAL vs continue PI/NNRTI. CONCLUSION In women with HIV and central obesity, the hepatic steatosis/fibrosis marker Chi3L1 and adiponectin decrease in conjunction with sST2 decreases following switch to RAL. Whether switching from NNRTI/PI-based regimens to RAL can improve hepatic steatosis and dysmetabolism requires further study. TRIAL REGISTRATION Clinicaltrials.gov NCT00656175.
Collapse
Affiliation(s)
- Obiageli Offor
- Department of Epidemiology, University of Texas Health Science Center Houston, Texas, United States of America
| | - Netanya Utay
- Department of Internal Medicine, University of Texas Health Science Center Houston, Texas, United States of America
| | - David Reynoso
- Department of Infectious Disease, University of Texas Medical Branch Galveston, Texas, United States of America
| | - Anoma Somasunderam
- Department of Internal Medicine, University of Texas Health Science Center Houston, Texas, United States of America
| | - Judith Currier
- Department of Infectious Disease, University of California Los Angeles, California, United States of America
| | - Jordan Lake
- Department of Infectious Disease, University of Texas Health Science Center Houston, Texas, United States of America
| |
Collapse
|
21
|
Carson JP, Ramm GA, Robinson MW, McManus DP, Gobert GN. Schistosome-Induced Fibrotic Disease: The Role of Hepatic Stellate Cells. Trends Parasitol 2018. [PMID: 29526403 DOI: 10.1016/j.pt.2018.02.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hepatic fibrosis is a common pathology in various liver diseases. Hepatic stellate cells (HSCs) are the main cell type responsible for collagen deposition and fibrosis formation in the liver. Schistosomiasis is characterised by granulomatous fibrosis around parasite eggs trapped within the liver and other host tissues. This response is facilitated by the recruitment of immune cells and the activation of HSCs. The interactions between HSCs and schistosome eggs are complex and diverse, and a better understanding of these interactions could lead to improved resolution of fibrotic liver disease, including that associated with schistosomiasis. Here, we discuss recent advances in HSC biology and the role of HSCs in hepatic schistosomiasis.
Collapse
Affiliation(s)
- Jack P Carson
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Locked Bag 2000, Royal Brisbane Hospital QLD 4029, Australia; Faculty of Medicine, The University of Queensland, Level 6, Oral Health Centre (Building), Herston Road, Herston, QLD, 4006, Australia
| | - Mark W Robinson
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Donald P McManus
- QIMR Berghofer Medical Research Institute, Locked Bag 2000, Royal Brisbane Hospital QLD 4029, Australia
| | - Geoffrey N Gobert
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
22
|
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by inflammation and fibrosis, in addition to steatosis, of the liver, but no therapeutic agents have yet been established. The mast cell protease chymase can generate angiotensin II, matrix metalloproteinase-9 and transforming growth factor-β, all of which are associated with liver inflammation or fibrosis. In animal models of NASH, augmented chymase has been observed in the liver. In histological analysis, chymase inhibitor prevented hepatic steatosis, inflammation, and fibrosis. Chymase inhibitor also attenuated the augmentation of angiotensin II, matrix metalloproteinase-9, and transforming growth factor-β observed in the liver of NASH. Oxidative stress, inflammatory markers, and collagen were attenuated by chymase inhibition. Moreover, chymase inhibitor showed a mitigating effect on established NASH, and survival rates were significantly increased by treatment with chymase inhibitor. In this review, we propose that chymase inhibitor has potential as a novel therapy for NASH.
Collapse
Affiliation(s)
- Shinji Takai
- Department of Innovative Medicine, Graduate School of Medicine, Osaka Medical College, Takatsuki, Japan
| | - Denan Jin
- Department of Innovative Medicine, Graduate School of Medicine, Osaka Medical College, Takatsuki, Japan
| |
Collapse
|
23
|
Wu T, Zhang Q, Song H. Swertiamarin attenuates carbon tetrachloride (CCl4)-induced liver injury and inflammation in rats by regulating the TLR4 signaling pathway. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000417449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Tao Wu
- Huazhong University of Science and Technology, China
| | - Qianrui Zhang
- General Hospital of the Yangtze River Shipping, China
| | - Hongping Song
- Huazhong University of Science and Technology, China
| |
Collapse
|
24
|
Cai Y, Huang G, Ma L, Dong L, Chen S, Shen X, Zhang S, Xue R, Sun D, Zhang S. Smurf2, an E3 ubiquitin ligase, interacts with PDE4B and attenuates liver fibrosis through miR-132 mediated CTGF inhibition. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:297-308. [PMID: 29100790 DOI: 10.1016/j.bbamcr.2017.10.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/23/2017] [Accepted: 10/29/2017] [Indexed: 12/13/2022]
Abstract
We previously reported that Smad ubiquitin regulatory factor 2 (Smurf2) activity was decreased in human fibrotic livers. Here, we overexpressed Smurf2 in livers of transgenic mice and observed inhibited collagen deposition and hepatic stellate cell activation in fibrotic model induced by carbon tetrachloride treatment or bile duct ligation. Hepatic Smurf2 overexpression also inhibited the production of connective tissue growth factor (CTGF), a central mediator of liver fibrosis. Using miRNA array and bioinformatics analyses, we identified miR-132 as a mediator of this inhibitory effect. miR-132 directly targets the 3'-untranslated region of CTGF and was transcriptionally upregulated by cAMP-PKA-CREB signaling. In addition, Smurf2 activated cAMP-PKA-CREB pathway by interacting with phosphodiesterase 4B (PDE4B) and facilitating its degradation. Thus, we have demonstrated a previously unrecognized anti-fibrotic pathway controlled by Smurf2.
Collapse
Affiliation(s)
- Yu Cai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Disease, Fudan University, Shanghai, China
| | - Guanqun Huang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| | - Lijie Ma
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Disease, Fudan University, Shanghai, China
| | - She Chen
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Disease, Fudan University, Shanghai, China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Disease, Fudan University, Shanghai, China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Disease, Fudan University, Shanghai, China.
| | - Deqiang Sun
- Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX, USA.
| | - Si Zhang
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Enhanced Wnt Signalling in Hepatocytes is Associated with Schistosoma japonicum Infection and Contributes to Liver Fibrosis. Sci Rep 2017; 7:230. [PMID: 28331224 PMCID: PMC5428310 DOI: 10.1038/s41598-017-00377-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 02/23/2017] [Indexed: 12/16/2022] Open
Abstract
Liver fibrosis is the most serious pathology caused by Schistosoma japonicum infection, which arises when schistosome eggs are deposited in the liver. Eosinophils, macrophages and hepatic stellate cells (HSCs) have been identified as major cellular contributors to the development of granulomas and fibrosis, but little is known about the effects of hepatocytes on granuloma formation. Here, we found that the levels of Wnt signalling-related molecules, transforming growth factor β (TGF-β) and connective tissue growth factor (CTGF) in hepatocytes were markedly elevated after S. japonicum infection. Liver fibrosis was exacerbated when exogenous Wnt3a was introduced, but was alleviated when Wnt signalling was suppressed by DKK1, accompanied by the reduced expression of TGF-β and CTGF in hepatocytes. These results indicate that the hepatocytic expression of TGF-β and CTGF is mediated by Wnt signalling. Additionally, the hepatocytes isolated from infected mice promoted the activation of primary HSCs in vitro, however, this effect was not observed when hepatocytes from DKK1 treated S. japonicum-infected mice was employed in the co-culture system. Our findings identify a novel pro-fibrogenic role of hepatocytes in schistosomiasis-induced liver fibrosis that is dependent on Wnt signalling, which may serve as a potential target for ameliorating hepatic fibrosis caused by helminths.
Collapse
|
26
|
Amygdalin inhibits HSC-T6 cell proliferation and fibrosis through the regulation of TGF-β/CTGF. Mol Cell Toxicol 2016. [DOI: 10.1007/s13273-016-0031-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
27
|
Chen L, Chen R, Velazquez VM, Brigstock DR. Fibrogenic Signaling Is Suppressed in Hepatic Stellate Cells through Targeting of Connective Tissue Growth Factor (CCN2) by Cellular or Exosomal MicroRNA-199a-5p. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2921-2933. [PMID: 27662798 DOI: 10.1016/j.ajpath.2016.07.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/27/2016] [Accepted: 07/14/2016] [Indexed: 12/18/2022]
Abstract
Pathways of liver fibrosis are controlled by connective tissue growth factor (CCN2). In this study, CCN2 was identified as a target of miR-199a-5p, which was principally expressed in quiescent mouse hepatic stellate cells (HSCs) and directly suppressed production of CCN2. Up-regulated CCN2 expression in fibrotic mouse livers or in activated primary mouse HSCs was associated with miR-199a-5p down-regulation. MiR-199a-5p in quiescent mouse HSCs inhibited the activity of a wild-type CCN2 3' untranslated region (3'-UTR) but not of a mutant CCN2 3'-UTR lacking the miR-199a-5p-binding site. In activated mouse HSCs, CCN2, α-smooth muscle actin, and collagen 1(α1) were suppressed by a miR-199a-5p mimic, whereas in quiescent mouse HSCs, the inhibited CCN2 3'-UTR activity was blocked by a miR-199a-5p antagomir. CCN2 3'-UTR activity in human HSCs was reduced by a miR-199a-5p mimic. MiR-199a-5p was present at higher levels in exosomes from quiescent versus activated HSCs. MiR-199a-5p-containing exosomes were shuttled from quiescent mouse HSCs to activated mouse HSCs in which CCN2 3'-UTR activity was then suppressed. Exosomes from quiescent HSCs caused miR-199a-5p-dependent inhibition of CCN2, α-smooth muscle actin, or collagen 1(α1) in activated HSCs in vitro and bound to activated HSCs in vivo. Thus, CCN2 suppression by miR-199a-5p accounts, in part, for low-level fibrogenic gene expression in quiescent HSCs and causes dampened gene expression in activated HSCs after horizontal transfer of miR-199a-5p in exosomes from quiescent HSCs.
Collapse
Affiliation(s)
- Li Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Ruju Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Victoria M Velazquez
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - David R Brigstock
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Department of Surgery, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
28
|
Horvat T, Landesmann B, Lostia A, Vinken M, Munn S, Whelan M. Adverse outcome pathway development from protein alkylation to liver fibrosis. Arch Toxicol 2016; 91:1523-1543. [PMID: 27542122 PMCID: PMC5364266 DOI: 10.1007/s00204-016-1814-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
In modern toxicology, substantial efforts are undertaken to develop alternative solutions for in vivo toxicity testing. The adverse outcome pathway (AOP) concept could facilitate knowledge-based safety assessment of chemicals that does not rely exclusively on in vivo toxicity testing. The construction of an AOP is based on understanding toxicological processes at different levels of biological organisation. Here, we present the developed AOP for liver fibrosis and demonstrate a linkage between hepatic injury caused by chemical protein alkylation and the formation of liver fibrosis, supported by coherent and consistent scientific data. This long-term process, in which inflammation, tissue destruction, and repair occur simultaneously, results from the complex interplay between various hepatic cell types, receptors, and signalling pathways. Due to the complexity of the process, an adequate liver fibrosis cell model for in vitro evaluation of a chemical's fibrogenic potential is not yet available. Liver fibrosis poses an important human health issue that is also relevant for regulatory purposes. An AOP described with enough mechanistic detail might support chemical risk assessment by indicating early markers for downstream events and thus facilitating the development of an in vitro testing strategy. With this work, we demonstrate how the AOP framework can support the assembly and coherent display of distributed mechanistic information from the literature to support the use of alternative approaches for prediction of toxicity. This AOP was developed according to the guidance document on developing and assessing AOPs and its supplement, the users' handbook, issued by the Organisation for Economic Co-operation and Development.
Collapse
Affiliation(s)
- Tomislav Horvat
- Chemicals Safety and Alternative Methods Unit (F.3), Directorate F - Health, Consumers and Reference Materials, Directorate General Joint Research Centre, European Commission, Ispra, Italy
| | - Brigitte Landesmann
- Chemicals Safety and Alternative Methods Unit (F.3), Directorate F - Health, Consumers and Reference Materials, Directorate General Joint Research Centre, European Commission, Ispra, Italy.
| | - Alfonso Lostia
- Chemicals Safety and Alternative Methods Unit (F.3), Directorate F - Health, Consumers and Reference Materials, Directorate General Joint Research Centre, European Commission, Ispra, Italy
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Center for Pharmaceutical Research, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Sharon Munn
- Chemicals Safety and Alternative Methods Unit (F.3), Directorate F - Health, Consumers and Reference Materials, Directorate General Joint Research Centre, European Commission, Ispra, Italy
| | - Maurice Whelan
- Chemicals Safety and Alternative Methods Unit (F.3), Directorate F - Health, Consumers and Reference Materials, Directorate General Joint Research Centre, European Commission, Ispra, Italy
| |
Collapse
|
29
|
PAK proteins and YAP-1 signalling downstream of integrin beta-1 in myofibroblasts promote liver fibrosis. Nat Commun 2016; 7:12502. [PMID: 27535340 DOI: 10.1038/ncomms12502] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/06/2016] [Indexed: 12/14/2022] Open
Abstract
Fibrosis due to extracellular matrix (ECM) secretion from myofibroblasts complicates many chronic liver diseases causing scarring and organ failure. Integrin-dependent interaction with scar ECM promotes pro-fibrotic features. However, the pathological intracellular mechanism in liver myofibroblasts is not completely understood, and further insight could enable therapeutic efforts to reverse fibrosis. Here, we show that integrin beta-1, capable of binding integrin alpha-11, regulates the pro-fibrotic phenotype of myofibroblasts. Integrin beta-1 expression is upregulated in pro-fibrotic myofibroblasts in vivo and is required in vitro for production of fibrotic ECM components, myofibroblast proliferation, migration and contraction. Serine/threonine-protein kinase proteins, also known as P21-activated kinase (PAK), and the mechanosensitive factor, Yes-associated protein 1 (YAP-1) are core mediators of pro-fibrotic integrin beta-1 signalling, with YAP-1 capable of perpetuating integrin beta-1 expression. Pharmacological inhibition of either pathway in vivo attenuates liver fibrosis. PAK protein inhibition, in particular, markedly inactivates the pro-fibrotic myofibroblast phenotype, limits scarring from different hepatic insults and represents a new tractable therapeutic target for treating liver fibrosis.
Collapse
|
30
|
ω-3 PUFAs ameliorate liver fibrosis and inhibit hepatic stellate cells proliferation and activation by promoting YAP/TAZ degradation. Sci Rep 2016; 6:30029. [PMID: 27435808 PMCID: PMC4951777 DOI: 10.1038/srep30029] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/28/2016] [Indexed: 12/15/2022] Open
Abstract
Elevated levels of the transcriptional regulators Yes-associated protein (YAP) and transcriptional coactivators with PDZ-binding motif (TAZ), key effectors of the Hippo pathway, have been shown to play essential roles in controlling liver cell fate and the activation of hepatic stellate cells (HSCs). The dietary intake of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) has been positively associated with a number of health benefits including prevention and reduction of cardiovascular diseases, inflammation and cancers. However, little is known about the impact of ω-3 PUFAs on liver fibrosis. In this study, we used CCl4-induced liver fibrosis mouse model and found that YAP/TAZ is over-expressed in the fibrotic liver and activated HSCs. Fish oil administration to the model mouse attenuates CCl4-induced liver fibrosis. Further study revealed that ω-3 PUFAs down-regulate the expression of pro-fibrogenic genes in activated HSCs and fibrotic liver, and the down-regulation is mediated via YAP, thus identifying YAP as a target of ω-3 PUFAs. Moreover, ω-3 PUFAs promote YAP/TAZ degradation in a proteasome-dependent manner. Our data have identified a mechanism of ω-3 PUFAs in ameliorating liver fibrosis.
Collapse
|
31
|
TGF-β signaling is activated in patients with chronic HBV infection and repressed by SMAD7 overexpression after successful antiviral treatment. Inflamm Res 2016; 65:355-65. [PMID: 26856334 DOI: 10.1007/s00011-016-0921-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 11/07/2015] [Accepted: 01/27/2016] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES Although animal studies demonstrated that Smad7 induction ameliorates TGF-β/SMAD-mediated fibrogenesis, its role in human hepatic diseases is rather obscure. Our study explored the activation status of TGF-β/activin pathway in patients with chronic liver diseases, and how it is affected by successful antiviral treatment in chronic HBV hepatitis (CHB). METHODS Thirty-seven CHB patients (19 with active disease, 14 completely remitted on long-term antiviral treatment and 4 with relapse after treatment withdrawal), 18 patients with chronic HCV hepatitis, 12 with non-alcoholic fatty liver disease (NAFLD), and 3 controls were enrolled in the study. Liver mRNA levels of CTGF, all TGF-β/activin isoforms, their receptors and intracellular mediators (SMADs) were evaluated using qRT-PCR and were correlated with the grade of liver inflammation and fibrosis staging. The expression and localization of pSMAD2 and pSMAD3 were assessed by immunohistochemistry. RESULTS TGF-β signalling is activated in CHB patients with active disease, while SMAD7 is up-regulated during the resolution of inflammation after successful treatment. SMAD7 overexpression was also observed in NAFLD patients exhibiting no or minimal fibrosis, despite the activation of TGF-β/activin signaling. CONCLUSIONS SMAD7 overexpression might represent a mechanism limiting TGF-β-mediated fibrogenesis in human hepatic diseases; therefore, SMAD7 induction likely represents a candidate for novel therapeutic approaches.
Collapse
|
32
|
Kim KE, Kim H, Heo RW, Shin HJ, Yi CO, Lee DH, Kim HJ, Kang SS, Cho GJ, Choi WS, Roh GS. Myeloid-specific SIRT1 Deletion Aggravates Hepatic Inflammation and Steatosis in High-fat Diet-fed Mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:451-60. [PMID: 26330758 PMCID: PMC4553405 DOI: 10.4196/kjpp.2015.19.5.451] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/04/2015] [Accepted: 05/31/2015] [Indexed: 01/23/2023]
Abstract
Sirtuin 1 (SIRT1) is a mammalian NAD+-dependent protein deacetylase that regulates cellular metabolism and inflammatory response. The organ-specific deletion of SIRT1 induces local inflammation and insulin resistance in dietary and genetic obesity. Macrophage-mediated inflammation contributes to insulin resistance and metabolic syndrome, however, the macrophage-specific SIRT1 function in the context of obesity is largely unknown. C57/BL6 wild type (WT) or myeloid-specific SIRT1 knockout (KO) mice were fed a high-fat diet (HFD) or normal diet (ND) for 12 weeks. Metabolic parameters and markers of hepatic steatosis and inflammation in liver were compared in WT and KO mice. SIRT1 deletion enhanced HFD-induced changes on body and liver weight gain, and increased glucose and insulin resistance. In liver, SIRT1 deletion increased the acetylation, and enhanced HFD-induced nuclear translocation of nuclear factor kappa B (NF-κB), hepatic inflammation and macrophage infiltration. HFD-fed KO mice showed severe hepatic steatosis by activating lipogenic pathway through sterol regulatory element-binding protein 1 (SREBP-1), and hepatic fibrogenesis, as indicated by induction of connective tissue growth factor (CTGF), alpha-smooth muscle actin (α-SMA), and collagen secretion. Myeloid-specific deletion of SIRT1 stimulates obesity-induced inflammation and increases the risk of hepatic fibrosis. Targeted induction of macrophage SIRT1 may be a good therapy for alleviating inflammation-associated metabolic syndrome.
Collapse
Affiliation(s)
- Kyung Eun Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Hwajin Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Rok Won Heo
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Hyun Joo Shin
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Chin-Ok Yi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Dong Hoon Lee
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Wan Sung Choi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| |
Collapse
|
33
|
Zhou J, Liang Y, Pan JX, Wang FF, Lin XM, Ma RJ, Qu F, Fang JQ. Protein extracts of Crassostrea gigas alleviate CCl₄-induced hepatic fibrosis in rats by reducing the expression of CTGF, TGF-β1 and NF-κB in liver tissues. Mol Med Rep 2014; 11:2913-20. [PMID: 25434425 DOI: 10.3892/mmr.2014.3019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 05/09/2014] [Indexed: 11/05/2022] Open
Abstract
Hepatic fibrosis may contribute to liver carcinoma and the mortality of patients with hepatic fibrosis is gradually increasing. However, no definitive treatment has been established for hepatic fibrosis. The hepatic fibrotic process is reversible and can be controlled; therefore, the creation of novel and effective therapeutic methods to prevent or reverse the disease is required. The aim of the present study was to identify whether protein extracts from Pacific oysters (PEPO) could alleviate the hepatic fibrosis induced by CCl4 and to examine the mechanisms involved. A total of sixty rats were randomly divided into the following experimental groups: The normal control group; the hepatic fibrosis model group; the high‑dose; medium‑dose; and low‑dose PEPO groups; and the colchicine group. The results indicated that compared with those of the model group, PEPO treatment significantly decreased the serum levels of alanine aminotransferase, aspartate aminotransferase, γ‑glutamyltransferase, alkaline phosphatase, hyaluronic acid, laminin, collagen type IV and procollagen III in rats with hepatic fibrosis. The hematoxylin and eosin staining demonstrated that PEPO markedly alleviated hepatic fibrosis. The experiments using immunohistochemistry, western blotting and quantitative PCR indicated that protein and mRNA expression levels of connective tissue growth factor (CTGF), transforming growth factor β1 (TGFβ‑1) and nuclear factor κB (NF‑κB) in the liver tissues were significantly reduced by PEPO treatment. Therefore, it was concluded that PEPO successfully alleviated hepatic fibrosis induced by CCl4 and reversed the effects of hepatotoxicity by regulating the serum levels of enzymes and decreasing the expression levels of CTGF, TGF‑β1 and NF‑κB in liver tissues. These findings may provide a novel treatment option for patients with hepatic fibrosis in the future.
Collapse
Affiliation(s)
- Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310012, P.R. China
| | - Yi Liang
- Acupuncture Department, The Third Clinical College of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang 310035, P.R. China
| | - Jie-Xue Pan
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Fang-Fang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xian-Ming Lin
- Acupuncture Department, The Third Clinical College of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang 310035, P.R. China
| | - Rui-Jie Ma
- Acupuncture Department, The Third Clinical College of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang 310035, P.R. China
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Jian-Qiao Fang
- Acupuncture Department, The Third Clinical College of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang 310035, P.R. China
| |
Collapse
|
34
|
Kim S, Jung J, Kim H, Heo RW, Yi CO, Lee JE, Jeon BT, Kim WH, Hahm JR, Roh GS. Exendin-4 Improves Nonalcoholic Fatty Liver Disease by Regulating Glucose Transporter 4 Expression in ob/ob Mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:333-9. [PMID: 25177166 PMCID: PMC4146636 DOI: 10.4196/kjpp.2014.18.4.333] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 05/14/2014] [Accepted: 06/14/2014] [Indexed: 01/14/2023]
Abstract
Exendin-4 (Ex-4), a glucagon-like peptide-1 receptor (GLP-1R) agonist, has been known to reverse hepatic steatosis in ob/ob mice. Although many studies have evaluated molecular targets of Ex-4, its mechanism of action on hepatic steatosis and fibrosis has not fully been determined. In the liver, glucose transporter 4 (GLUT4) is mainly expressed in hepatocytes, endothelial cells and hepatic stellate cells (HSCs). In the present study, the effects of Ex-4 on GLUT4 expression were determined in the liver of ob/ob mice. Ob/ob mice were treated with Ex-4 for 10 weeks. Serum metabolic parameters, hepatic triglyceride levels, and liver tissues were evaluated for hepatic steatosis. The weights of the whole body and liver in ob/ob mice were reduced by long-term Ex-4 treatment. Serum metabolic parameters, hepatic steatosis, and hepatic fibrosis in ob/ob mice were reduced by Ex-4. Particularly, Ex-4 improved hepatic steatosis by enhancing GLUT4 via GLP-1R activation in ob/ob mice. Ex-4 treatment also inhibited hepatic fibrosis by decreasing expression of connective tissue growth factor in HSCs of ob/ob mice. Our data suggest that GLP-1 agonists exert a protective effect on hepatic steatosis and fibrosis in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Seok Kim
- Department of Anatomy and Convergence Medical Science, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| | - Jaehoon Jung
- Department of Internal Medicine, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| | - Hwajin Kim
- Department of Anatomy and Convergence Medical Science, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| | - Rok Won Heo
- Department of Anatomy and Convergence Medical Science, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| | - Chin-Ok Yi
- Department of Anatomy and Convergence Medical Science, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| | - Jung Eun Lee
- Department of Thoracic and Cardiovascular Surgery, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| | - Byeong Tak Jeon
- Department of Neurologic Surgery, Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Won-Ho Kim
- Division of Metabolic Diseases, Center for Biomedical Sciences, National Institutes of Health, Cheongwon-gun 363-700, Korea
| | - Jong Ryeal Hahm
- Department of Internal Medicine, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institutes of Health Science, Gyeongsnag National University School of Medicine, Jinju 660-751, Korea
| |
Collapse
|
35
|
Pan PH, Lin SY, Wang YY, Chen WY, Chuang YH, Wu CC, Chen CJ. Protective effects of rutin on liver injury induced by biliary obstruction in rats. Free Radic Biol Med 2014; 73:106-16. [PMID: 24815012 DOI: 10.1016/j.freeradbiomed.2014.05.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/30/2014] [Accepted: 05/01/2014] [Indexed: 12/14/2022]
Abstract
Rutin has been shown to possess beneficial health effects, including hepatoprotection. However, to date, it has not been demonstrated to have a hepatoprotective effect against cholestatic liver injury. This is the first report to show a protective effect of rutin on cholestatic liver injury. Cholestasis was produced by bile duct ligation (BDL) in male Sprague-Dawley rats for 3 weeks. Daily oral administration of rutin was started 1 week before injury and was maintained for 4 weeks. In comparison with the control group, the BDL group showed liver injury as evidenced by histological changes and elevation in serum biochemicals, ductular reaction, fibrosis, inflammation, and oxidative stress. These pathophysiological changes were attenuated by rutin supplementation. Rutin alleviated BDL-induced transforming growth factor β1 (TGF-β1), interleukin-1β, connective tissue growth factor, and collagen expression. The antifibrotic effect of rutin was accompanied by reductions in α-smooth muscle actin-positive matrix-producing cells and Smad2/3 activity critical to the fibrogenic potential of TGF-β1. Rutin attenuated BDL-induced oxidative stress, leukocyte accumulation, NF-κB activation, and proinflammatory cytokine production. Further studies demonstrated an inhibitory effect of rutin on the redox-sensitive intracellular signaling molecule extracellular signal-regulated kinase (ERK). Rutin also attenuated BDL-induced reduction in NF-E2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and AMP-activated protein kinase (AMPK). Taken together, the beneficial effects of rutin were shown to be associated with antioxidative and anti-inflammatory effects as well as the downregulation of NF-κB and TGF-β/Smad signaling, probably via interference of ERK activation and/or enhancement of Nrf2, HO-1, and AMPK activity.
Collapse
Affiliation(s)
- Pin-Ho Pan
- Department of Pediatrics, Tungs' Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Shih-Yi Lin
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Ya-Yu Wang
- Division of Family Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Yu-Han Chuang
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Chih-Cheng Wu
- Department of Anesthesiology, and Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan; Institute of Biomedical Sciences, and National Chung Hsing University, Taichung 402, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; Center for General Education, Tunghai University, Taichung 407, Taiwan; Graduate School of Nursing, Hungkuang University, Taichung 433, Taiwan.
| |
Collapse
|
36
|
Lin SY, Wang YY, Chen WY, Chuang YH, Pan PH, Chen CJ. Beneficial effect of quercetin on cholestatic liver injury. J Nutr Biochem 2014; 25:1183-1195. [PMID: 25108658 DOI: 10.1016/j.jnutbio.2014.06.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 05/29/2014] [Accepted: 06/04/2014] [Indexed: 12/15/2022]
Abstract
Bile duct obstruction and subsequent cholestasis are associated with hepatocellular injury, cholangiocyte proliferation, stellate cell activation, Kupffer cell activation, oxidative stress, inflammation and fibrosis. Flavonoids have been shown to confer beneficial health effects, including hepatoprotection. However, the molecular mechanism of flavonoid-mediated hepatoprotection is incompletely understood. In this study, we report the protective effect of quercetin on cholestatic liver injury. Cholestasis was produced by bile duct ligation (BDL) in male Sprague-Dawley rats for 3 weeks. Daily oral administration of quercetin was started 1 week before injury and lasted for 4 weeks. In comparison with the control group, the BDL group showed liver injury, as evidenced by histological changes, and elevation in serum biochemicals, ductular reaction, fibrosis, inflammation and oxidative stress. These pathophysiological changes were attenuated by daily quercetin supplementation. Quercetin alleviated BDL-induced transforming growth factor beta-1 (TGF-β1), interleukin-1 beta, connective tissue growth factor and collagen expression. The antifibrotic effect of quercetin was accompanied by reductions in α-smooth muscle actin-positive matrix-producing cells and Smad 2/3 activity critical to the fibrogenic potential of TGF-β1. Quercetin also attenuated BDL-induced oxidative stress, leukocyte accumulation, nuclear factor (NF)-κB activation and proinflammatory cytokine production. Further studies demonstrated an inhibitory effect of quercetin on MyD88 and TGF-β-activated kinase-1 critical for linking toll-like receptor (TLR) and NF-κB. Taken together, the hepatoprotective, anti-inflammatory and antifibrotic effects of quercetin seem to be multifactorial. The beneficial effects of daily quercetin supplementation are associated with antioxidative and anti-inflammatory potential as well as down-regulation of NF-κB and TGF-β/Smad signaling, probably via interference with TLR signaling.
Collapse
Affiliation(s)
- Shih-Yi Lin
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung 407, Taiwan; School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Ya-Yu Wang
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; Division of Family Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Yu-Han Chuang
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Pin-Ho Pan
- Department of Pediatrics, Tungs' Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan; Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402, Taiwan; Center for General Education, Tunghai University, Taichung 407, Taiwan; Department of Nursing, HungKuang University, Taichung 433, Taiwan.
| |
Collapse
|
37
|
Charrier A, Chen R, Chen L, Kemper S, Hattori T, Takigawa M, Brigstock DR. Exosomes mediate intercellular transfer of pro-fibrogenic connective tissue growth factor (CCN2) between hepatic stellate cells, the principal fibrotic cells in the liver. Surgery 2014; 156:548-55. [PMID: 24882759 DOI: 10.1016/j.surg.2014.04.014] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 04/14/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Fibrogenic pathways in the liver are principally regulated by hepatic stellate cells (HSC), which produce and respond to fibrotic mediators such as connective tissue growth factor (CCN2). The aim of this study was to determine whether CCN2 is shuttled between HSC in membranous nanovesicles, or "exosomes." METHODS Exosomes were incubated with HSC after isolation from conditioned medium of control or CCN2-green fluorescent protein (GFP)-transfected primary mouse HSC or human LX-2 HSC. Some exosomes were stained fluorescently with PKH26. HSC co-culture experiments were performed in the presence of GW4869 exosome inhibitor. CCN2 or CCN2-GFP were evaluated by quantitative real-time polymerase chain reaction or Western blot. RESULTS HSC-derived exosomes contained CCN2 or CCN2 mRNA, each of which increased in concentration during HSC activation or after transfection of HSC with CCN2-GFP. Exosomes, stained with either PKH26 or purified from CCN2-GFP-transfected cells, were taken up by activated or quiescent HSC resulting in CCN2-GFP delivery, as shown by their direct addition to recipient cells or by the GW4869-dependency of donor HSC. CONCLUSION CCN2 is packaged into secreted, nano-sized exosomes that mediate its intercellular transfer between HSC. Exosomal CCN2 may amplify or fine tune fibrogenic signaling and, in conjunction with other exosome constituents, may have utility as a noninvasive biomarker to assess hepatic fibrosis.
Collapse
Affiliation(s)
- Alyssa Charrier
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Ruju Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Li Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Sherri Kemper
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Takako Hattori
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masaharu Takigawa
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School, Okayama, Japan
| | - David R Brigstock
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH; Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH; Department of Surgery, The Ohio State University, Columbus, OH.
| |
Collapse
|
38
|
Chen L, Charrier A, Zhou Y, Chen R, Yu B, Agarwal K, Tsukamoto H, Lee LJ, Paulaitis ME, Brigstock DR. Epigenetic regulation of connective tissue growth factor by MicroRNA-214 delivery in exosomes from mouse or human hepatic stellate cells. Hepatology 2014; 59:1118-29. [PMID: 24122827 PMCID: PMC3943742 DOI: 10.1002/hep.26768] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 09/20/2013] [Indexed: 12/14/2022]
Abstract
UNLABELLED Connective tissue growth factor (CCN2) drives fibrogenesis in hepatic stellate cells (HSC). Here we show that CCN2 up-regulation in fibrotic or steatotic livers, or in culture-activated or ethanol-treated primary mouse HSC, is associated with a reciprocal down-regulation of microRNA-214 (miR-214). By using protector or reporter assays to investigate the 3'-untranslated region (UTR) of CCN2 mRNA, we found that induction of CCN2 expression in HSC by fibrosis-inducing stimuli was due to reduced expression of miR-214, which otherwise inhibited CCN2 expression by directly binding to the CCN2 3'-UTR. Additionally, miR-214 was present in HSC exosomes, which were bi-membrane vesicles, 50-150 nm in diameter, negatively charged (-26 mV), and positive for CD9. MiR-214 levels in exosomes but not in cell lysates were reduced by pretreatment of the cells with the exosome inhibitor, GW4869. Coculture of either quiescent HSC or miR-214-transfected activated HSC with CCN2 3'-UTR luciferase reporter-transfected recipient HSC resulted in miR-214- and exosome-dependent regulation of a wild-type CCN2 3'-UTR reporter but not of a mutant CCN2 3'-UTR reporter lacking the miR-214 binding site. Exosomes from HSC were a conduit for uptake of miR-214 by primary mouse hepatocytes. Down-regulation of CCN2 expression by miR-214 also occurred in human LX-2 HSC, consistent with a conserved miR-214 binding site in the human CCN2 3'-UTR. MiR-214 in LX-2 cells was shuttled by way of exosomes to recipient LX-2 cells or human HepG2 hepatocytes, resulting in suppression of CCN2 3'-UTR activity or expression of CCN2 downstream targets, including alpha smooth muscle actin or collagen. Experimental fibrosis in mice was associated with reduced circulating miR-214 levels. CONCLUSION Exosomal transfer of miR-214 is a paradigm for the regulation of CCN2-dependent fibrogenesis and identifies fibrotic pathways as targets of intercellular regulation by exosomal miRs.
Collapse
Affiliation(s)
- Li Chen
- The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205
| | - Alyssa Charrier
- The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205,Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus OH 43212
| | - Yu Zhou
- The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205
| | - Ruju Chen
- The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205
| | - Bo Yu
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210,Nanoscale Science and Engineering Center, The Ohio State University, Columbus, OH 43210
| | - Kitty Agarwal
- Nanoscale Science and Engineering Center, The Ohio State University, Columbus, OH 43210,Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
| | - Hidekazu Tsukamoto
- Department of Pathology, Southern California Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089,Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073
| | - L. James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210,Nanoscale Science and Engineering Center, The Ohio State University, Columbus, OH 43210
| | - Michael E Paulaitis
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210,Nanoscale Science and Engineering Center, The Ohio State University, Columbus, OH 43210
| | - David R Brigstock
- The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205,Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus OH 43212,Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43212,Address Correspondence to: David Brigstock, Ph.D., Room 2011, Research Building 2, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205, Tel 614-355-2824,
| |
Collapse
|
39
|
Zhang F, Zhang Z, Kong D, Zhang X, Chen L, Zhu X, Lu Y, Zheng S. Tetramethylpyrazine reduces glucose and insulin-induced activation of hepatic stellate cells by inhibiting insulin receptor-mediated PI3K/AKT and ERK pathways. Mol Cell Endocrinol 2014; 382:197-204. [PMID: 24071517 DOI: 10.1016/j.mce.2013.09.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/05/2013] [Accepted: 09/17/2013] [Indexed: 01/19/2023]
Abstract
Hepatic stellate cell (HSC) activation is the central event during liver fibrogenesis. Metabolic syndrome characterized by hyperglycemia and hyperinsulinemia contributes to nonalcoholic steatohepatitis-associated liver fibrosis. This study was to investigate the effects of tetramethylpyrazine (TMP) on HSC activation induced by glucose and insulin (Glu/Ins) and the underlying mechanisms. Results showed that Glu/Ins significantly stimulated proliferation, invasion, adhesion, and extracellular matrix (ECM) production in HSCs. TMP inhibited HSC proliferation, invasion and adhesion, and reduced the expression of marker genes related to HSC activation in Glu/Ins-activated HSCs. Mechanistic evidence revealed that TMP reduced insulin receptor (InsR) expression and blocked the downstream phosphatidylinositol-3-kinase (PI3K)/AKT and extracellular signal-regulated kinase (ERK) cascades, which was required for TMP attenuation of HSC activation. Moreover, TMP modulated the genes relevant to ECM homeostasis favoring ECM degradation. It could be concluded that TMP inhibited Glu/Ins-stimulated HSC activation and ECM production by inhibiting InsR-mediated PI3K/AKT and ERK pathways.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zili Zhang
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Desong Kong
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoping Zhang
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Chen
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaojing Zhu
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yin Lu
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; The National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shizhong Zheng
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; The National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
40
|
Liu Y, Liu H, Meyer C, Li J, Nadalin S, Königsrainer A, Weng H, Dooley S, Ten Dijke P. Transforming growth factor-β (TGF-β)-mediated connective tissue growth factor (CTGF) expression in hepatic stellate cells requires Stat3 signaling activation. J Biol Chem 2013; 288:30708-30719. [PMID: 24005672 DOI: 10.1074/jbc.m113.478685] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In fibrotic liver, connective tissue growth factor (CTGF) is constantly expressed in activated hepatic stellate cells (HSCs) and acts downstream of TGF-β to modulate extracellular matrix production. Distinct from other cell types in which Smad signaling plays major role in regulating CTGF production, TGF-β stimulated CTGF expression in activated HSCs is only in part dependent on Smad3. Other signaling molecules like MAPKs and PI3Ks may also participate in this process, and the underlying mechanisms have yet to be clarified. In this study, we report involvement of Stat3 activation in modulating CTGF production upon TGF-β challenge in activated HSCs. Stat3 is phosphorylated via JAK1 and acts as a critical ALK5 (activin receptor-like kinase 5) downstream signaling molecule to mediate CTGF expression. This process requires de novo gene transcription and is additionally modulated by MEK1/2, JNK, and PI3K pathways. Cell-specific knockdown of Smad3 partially decreases CTGF production, whereas it has no significant influence on Stat3 activation. The total CTGF production induced by TGF-β in activated HSCs is therefore, to a large extent, dependent on the balance and integration of the canonical Smad3 and Stat3 signaling pathways.
Collapse
Affiliation(s)
- Yan Liu
- From the Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany,; the Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands and Centre for Biomedical Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Heng Liu
- From the Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany,; the Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Christoph Meyer
- From the Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Jun Li
- the Department of General, Visceral Surgery and Transplantation, University Hospital Tübingen, 72076 Tübingen, Germany, and; the Department of Hepatobiliary Surgery and Visceral Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Silvio Nadalin
- the Department of General, Visceral Surgery and Transplantation, University Hospital Tübingen, 72076 Tübingen, Germany, and
| | - Alfred Königsrainer
- the Department of General, Visceral Surgery and Transplantation, University Hospital Tübingen, 72076 Tübingen, Germany, and
| | - Honglei Weng
- From the Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Steven Dooley
- From the Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany,.
| | - Peter Ten Dijke
- the Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands and Centre for Biomedical Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands,.
| |
Collapse
|
41
|
Connective tissue growth factor level is increased in patients with liver cirrhosis but is not associated with complications or extent of liver injury. ACTA ACUST UNITED AC 2012; 179:10-4. [DOI: 10.1016/j.regpep.2012.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 07/23/2012] [Accepted: 08/27/2012] [Indexed: 01/20/2023]
|
42
|
Chen WY, Lin SY, Pan HC, Liao SL, Chuang YH, Yen YJ, Lin SY, Chen CJ. Beneficial effect of docosahexaenoic acid on cholestatic liver injury in rats. J Nutr Biochem 2012; 23:252-264. [PMID: 21497498 DOI: 10.1016/j.jnutbio.2010.11.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 11/09/2010] [Accepted: 11/23/2010] [Indexed: 01/20/2023]
Abstract
Bile duct obstruction and subsequent cholestasis are associated with hepatocellular injury, cholangiocyte proliferation, stellate cell activation, Kupffer cell activation, oxidative stress, inflammation and fibrosis. Docosahexaenoic acid (DHA) is an essential polyunsaturated fatty acid that has been shown to possess health beneficial effects, including hepatoprotection. However, the molecular mechanism of DHA-mediated hepatoprotection is not fully understood. In the present study, we report the protective effect of DHA on cholestatic liver injury. Cholestasis was produced by bile duct ligation (BDL) in male Sprague-Dawley rats for 3 weeks. Daily administration of DHA was started 2 weeks before injury and lasted for 5 weeks. In comparison with the control group, the BDL group showed hepatic damage as evidenced by histological changes and elevation in serum biochemicals, ductular reaction, fibrosis, inflammation and oxidative stress. These pathophysiological changes were attenuated by chronic DHA supplementation. DHA alleviated BDL-induced transforming growth factor beta-1 (TGF-β1), intereukin-1beta, connective tissue growth factor and collagen expression. The anti-fibrotic effect of DHA was accompanied by reductions in α-smooth muscle actin-positive matrix-producing cells and Smad 2/3 activity critical to the fibrogenic potential of TGF-β1. DHA also attenuated BDL-induced leukocyte accumulation and nuclear factor-κB (NF-κB) activation. Further studies demonstrated an inhibitory effect of DHA on redox-sensitive intracellular signaling molecule extracellular signal-regulated kinase (ERK). Taken together, the hepatoprotective, anti-inflammatory and anti-fibrotic effects of DHA seem to be multifactorial. The beneficial effects of chronic DHA supplementation are associated with anti-oxidative and anti-inflammatory potential as well as down-regulation of NF-κB and transforming growth factor beta/Smad signaling probably via interference with ERK activation.
Collapse
Affiliation(s)
- Wen-Ying Chen
- Department of Education and Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Chen L, Charrier AL, Leask A, French SW, Brigstock DR. Ethanol-stimulated differentiated functions of human or mouse hepatic stellate cells are mediated by connective tissue growth factor. J Hepatol 2011; 55:399-406. [PMID: 21156189 PMCID: PMC3136646 DOI: 10.1016/j.jhep.2010.11.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 11/10/2010] [Accepted: 11/15/2010] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Connective tissue growth factor (CTGF) expression is intimately associated with hepatic fibrotic pathophysiology. In this study, CTGF production and action was investigated in ethanol-treated mouse primary hepatic stellate cells (HSC) or human LX-2 cells. METHODS CTGF, transforming growth factor-beta1 (TGF-β1), alpha-smooth muscle actin (α-SMA) or collagen α1(I) mRNA were quantified by real-time PCR after treatment of HSC with ethanol or acetaldehyde. CTGF protein production was assessed by immunoprecipitation or ELISA. Ethanol-stimulated CTGF transcription was investigated using CTGF promoter reporter constructs. The TGF-β1- or CTGF-dependency of ethanol-induced CTGF, α-SMA, or collagen α1(I) was determined using small interfering RNA (siRNA) to TGF-β1 or CTGF. RESULTS In human steatohepatitis, CTGF was produced by presumptive activated HSC. In cultured human or mouse HSC, production of CTGF, α-SMA and/or collagen was increased by ethanol treatment, an effect mimicked by acetaldehyde and blocked by 4-methylpyrazole (4-MP) or N-acetylcysteine (NAC). CTGF promoter activity was stimulated in a sustained fashion by ethanol or TGF-β1. Mutation of the Smad site or basal control element (BCE-1) in the CTGF promoter caused a 5-fold reduction in ethanol-stimulated CTGF promoter activity. Administration of TGF-β1 siRNA or CTGF siRNA significantly decreased ethanol- or acetaldehyde-stimulated mRNA or protein levels of CTGF, α-SMA or collagen I in LX-2 cells. In mouse HSC, TGF-β1- or ethanol-stimulated CTGF, α-SMA or collagen I were significantly attenuated by CTGF siRNA. CONCLUSIONS Ethanol-induced α-SMA or collagen α1(I) in HSC are mediated via TGF-β-dependent CTGF production, highlighting potential therapeutic benefits of targeting CTGF in alcoholic liver disease.
Collapse
Affiliation(s)
- Li Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children’s Hospital, Columbus OH 43205
| | - Alyssa L. Charrier
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children’s Hospital, Columbus OH 43205, Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus OH 43212
| | - Andrew Leask
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, N6G 1G9 Canada
| | - Samuel W. French
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA 90509
| | - David R. Brigstock
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children’s Hospital, Columbus OH 43205, Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus OH 43212, Department of Surgery, The Ohio State University, Columbus, OH 43212,Address correspondence to: David R. Brigstock, Center for Clinical and Translational Research, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus OH 43205. Tel: 614-355-2824; Fax: 614-722-5892;
| |
Collapse
|
44
|
Kodama T, Takehara T, Hikita H, Shimizu S, Shigekawa M, Tsunematsu H, Li W, Miyagi T, Hosui A, Tatsumi T, Ishida H, Kanto T, Hiramatsu N, Kubota S, Takigawa M, Tomimaru Y, Tomokuni A, Nagano H, Doki Y, Mori M, Hayashi N. Increases in p53 expression induce CTGF synthesis by mouse and human hepatocytes and result in liver fibrosis in mice. J Clin Invest 2011; 121:3343-56. [PMID: 21747166 DOI: 10.1172/jci44957] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 05/12/2011] [Indexed: 12/16/2022] Open
Abstract
The tumor suppressor p53 has been implicated in the pathogenesis of non-cancer-related conditions such as insulin resistance, cardiac failure, and early aging. In addition, accumulation of p53 has been observed in the hepatocytes of individuals with fibrotic liver diseases, but the significance of this is not known. Herein, we have mechanistically linked p53 activation in hepatocytes to liver fibrosis. Hepatocyte-specific deletion in mice of the gene encoding Mdm2, a protein that promotes p53 degradation, led to hepatocyte synthesis of connective tissue growth factor (CTGF; the hepatic fibrogenic master switch), increased hepatocyte apoptosis, and spontaneous liver fibrosis; concurrent removal of p53 completely abolished this phenotype. Compared with wild-type controls, mice with hepatocyte-specific p53 deletion exhibited similar levels of hepatocyte apoptosis but decreased liver fibrosis and hepatic CTGF expression in two models of liver fibrosis. The clinical significance of these data was highlighted by two observations. First, p53 upregulated CTGF in a human hepatocellular carcinoma cell line by repressing miR-17-92. Second, human liver samples showed a correlation between CTGF and p53-regulated gene expression, which were both increased in fibrotic livers. This study reveals that p53 induces CTGF expression and promotes liver fibrosis, suggesting that the p53/CTGF pathway may be a therapeutic target in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Haafiz A, Farrington C, Andres J, Islam S. Relationship between hepatic CTGF expression and routine blood tests at the time of liver transplantation for biliary atresia: hope or hype for a biomarker of hepatic fibrosis. Clin Exp Gastroenterol 2011; 4:49-54. [PMID: 21694872 PMCID: PMC3108679 DOI: 10.2147/ceg.s17145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Indexed: 11/23/2022] Open
Abstract
Background: Progressive hepatic fibrosis (HF) is a prominent feature of biliary atresia (BA), the most common indication for liver transplantation (LT) in children. Despite its importance in BA, HF is not evaluated in routine patient care because the invasiveness of liver biopsy makes histologic monitoring of fibrosis unfeasible. Therefore, the identification of noninvasive markers to assess HF is desirable especially in children. Purpose: The main goal of this pilot project was to establish an investigational framework correlating hepatic expression of fibrogenic markers with routine blood tests in BA. Methods: Using liver explants from patients with BA (n = 26), immune-expression of connective tissue growth factor (CTGF), a key fibrogenic cytokine was determined using horseradish-labeled antibodies. Expression intensities of lobular (L-CTGF) and portal (P-CTGF) CTGF were determined by using ImageJ software. These CTGF intensities were correlated with blood tests performed at the time of LT. Correlation coefficients were determined for each blood test variable versus mean L-CTGF and P-CTGF expression intensities. A P-value of less than 0.05 was considered statistically significant. Results: All patients had end-stage liver disease and persistent cholestasis at the time of LT. Kendall tau (τ) rank correlation coefficient for L-CTGF and white blood cell (WBC) was inversed (−0.52; P ≤ 0.02). Similar but statistically nonsignificant inverse relationships were noted between L-CTGF and prothrombin time (PT) (−0.15; P ≤ 0.4), international normalized ratio (INR) (−0.14; P ≤ 0.5), and platelet count (−0.36; P ≤ 0.09). Inversed (τ) rank correlation coefficients were also evident between P-CTGF expression and gamma-glutamyl transpeptidase (GGT), PT, INR, and platelet count. Pearson correlation coefficients for combinational analysis of standardized total bilirubin (TB), alkaline phosphatase, GGT, and platelet count with L-CTGF (0.33; P = 0.3) and P-CTGF (0.06; P = 0.8), were not significant. Similar analysis for alanine aminotransferase, TB, and GGT combination (L-CTGF, 0.16; P = 0.5; P-CTGF −0.3; P = 0.2) as well as WBC, platelet count, and TB (L-CTGF: −0.36; P = 0.09; P-CTGF −0.33; P = 0.13) also revealed nonsignificant results. Conclusion: Hepatic expression of fibrogenic markers can be correlated with routinely performed blood tests in patients with BA. We document that although a trend of inverse relationship is noted, hepatic CTGF expression does not correlate well with routinely performed blood tests in advanced BA. Further work is required to determine more reliable ways of noninvasive diagnosis of HF.
Collapse
Affiliation(s)
- Allah Haafiz
- Hepatology and Liver Transplantation, Division of Pediatric Gastroenterology, Hepatology and Nutrition
| | | | | | | |
Collapse
|
46
|
Farrington C, Novak D, Liu C, Haafiz AB. Immunohistochemical localization of transforming growth factor β-1 and its relationship with collagen expression in advanced liver fibrosis due to biliary atresia. Clin Exp Gastroenterol 2010; 3:185-91. [PMID: 21694865 PMCID: PMC3108674 DOI: 10.2147/ceg.s14220] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Biliary atresia (BA) is the most common indication of liver transplantation in children. Pathogenesis of hepatic fibrosis, which is a prominent feature of BA, remains obscure. The purpose of this work was to determine the cellular sources of transforming growth factor beta-1 (TGFβ1) and establish the relationship between TGFβ1-producing cells and extracellular matrix producing myofibroblasts (MFBs) in advanced BA. METHODS Trichrome staining and immunohistochemistry were carried out to determine the expression pattern of collagen and TGFβ1 protein in explant liver specimens from patients with BA. The intensities of portal and lobular TGFβ1 expressions were compared. Immunofluorescence technique was carried out to determine the relationship between α-smooth muscle actin (α-SMA)-positive-MFB and TGFβ1-positve cells. RESULTS Lobular TGFβ1 protein expression was significantly higher than portal (89 ± 6 versus 10 ± 1 arbitrary units, P ≤ 0.05), whereas no difference was noted in livers used as control (10 ± 1.6 versus 19 ± 5 arbitrary units, P = 0.11). TGFβ1 expression was more in the center of nodules versus MFB in surrounding fibrous septa. Contrary to TGFβ1 expression, α1-SMA was mostly expressed in the portal structures and the adjacent fibrous septa enacting lobulation of the parenchyma. The results obtained by coimmunofluorescence staining showed no colocalization of α-SMA and TGFβ1. CONCLUSIONS TGFβ1 protein expression is mostly localized to hepatocytes in advanced BA. These findings suggest a paracrine mechanisms of TGFβ1-driven fibrogenesis in advanced BA.
Collapse
Affiliation(s)
- Christian Farrington
- Hepatology and Liver Transplantation, Division of Pediatric Gastroenterology, Hepatology and Nutrition
| | | | | | | |
Collapse
|
47
|
Abstract
Hepatic steatosis is commonly seen in patients with chronic hepatitis C infection, and the two together have a greater association than by chance alone. Hepatitis C virus is closely associated with lipid metabolism throughout its lifecycle. Hepatic steatosis is more common in genotype 3 infection, due to direct viral effects including through microsomal triglyceride transfer protein, peroxisome proliferator activating receptor, and sterol regulatory element binding protein. In non-genotype 3 infection, hepatic steatosis is considered largely to be due to alterations in host metabolism, particularly through insulin resistance. The clinical relevance of this association has yet to be fully explored. Hepatic steatosis is associated with increased hepatic fibrosis and a reduced level of sustained virological response to pegylated interferon and ribavirin. Small studies trialing adjuvant anti-diabetic therapies or HMG-CoA reductase inhibitors with pegylated-interferon and ribavirin have shown an improved sustained virological response and reduced viral titer. Furthermore, simple lifestyle alterations showed positive effects on parameters of disease activity. These insights raise the possibility of novel treatment options.
Collapse
Affiliation(s)
- J H Patel
- Liver Unit, Imperial College London, St Mary's Hospital Campus, 10th Floor QEQM Building, Praed Street, London W2 1NY, UK
| | | | | | | |
Collapse
|
48
|
Huang G, Brigstock DR. Integrin expression and function in the response of primary culture hepatic stellate cells to connective tissue growth factor (CCN2). J Cell Mol Med 2010; 15:1087-95. [PMID: 20406330 PMCID: PMC2912974 DOI: 10.1111/j.1582-4934.2010.01072.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Production of connective tissue growth factor (CCN2, also known as CTGF) is a hallmark of hepatic fibrosis. This study examined early primary cultures of hepatic stellate cells (HSC) for (i) CCN2 regulation of its cognate receptor integrin subunits; and (ii) interactions between CCN2 and integrin α(5)β(1), heparan sulphate proteoglycans (HSPG) or fibronectin (FN) in supporting cell adhesion. HSC were isolated from healthy male Balb/c mice. mRNA levels of CCN2 or α(5), β(1), αv or β(3) integrin subunits were measured in days 1-7 primary culture HSC, and day 3 or day 7 cells treated with recombinant CCN2 or CCN2 small interfering RNA. Interactions between CCN2 and integrin α(5)β(1), HSPG or FN were investigated using an in vitro cell adhesion assay. Co-incident with autonomous activation over the first 7 days, primary culture HSC increasingly expressed mRNA for CCN2 or integrin subunits. Addition of exogenous CCN2 or knockdown of endogenous CCN2 differentially regulated integrin gene expression in day 3 versus day 7 cells. Either full length CCN2 ('CCN2(1-4)') or residues 247-349 containing module 4 alone ('CCN2(4)') supported day 3 cell adhesion in an integrin α(5)β(1) - and HSPG-dependent fashion. Adhesion of day 3 cells to FN was promoted in an integrin α(5) β(1)-dependent manner by CCN2(1-4) or CCN2(4), whereas FN promoted HSPG-dependent HSC adhesion to CCN2(1-4) or CCN2(4). These findings suggest CCN2 regulates integrin expression in primary culture HSC and supports HSC adhesion via its binding of cell surface integrin α(5)β(1), a novel CCN2 receptor in primary culture HSC which interacts co-operatively with HSPG or FN.
Collapse
Affiliation(s)
- Guangcun Huang
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | |
Collapse
|
49
|
Li GS, Jiang WL, Tian JW, Qu GW, Zhu HB, Fu FH. In vitro and in vivo antifibrotic effects of rosmarinic acid on experimental liver fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2010; 17:282-8. [PMID: 19524418 DOI: 10.1016/j.phymed.2009.05.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 04/03/2009] [Accepted: 05/05/2009] [Indexed: 05/27/2023]
Abstract
This study was carried out to investigate whether rosmarinic acid (RA) has antifibrotic effect on experimental liver fibrosis in vitro and in vivo and its possible mechanism. Culture of hepatic stellate cells (HSCs) determine proliferation and expression of transforming growth factor-beta1 (TGF-beta1), connective transforming growth factor (CTGF) and alpha-smooth muscle actin (alpha-SMA). In carbon tetrachloride (CCL(4))-induced rat liver fibrosis model, determined biochemical indicator, liver fibrosis grade and histopathological changes, immunohistochemical detected liver TGF-beta1 and CTGF expression. The results indicated that RA could inhibit HSCs proliferation, inhibit TGF-beta1, CTGF and alpha-SMA expression in cultured HSCs. It has marked evident in reducing fibrosis grade, ameliorating biochemical indicator and histopathological morphology, reducing liver TGF-beta1 and CTGF expression in CCL(4)-induced liver fibrosis. These findings suggest that RA has potentially conferring antifibrogenic effects.
Collapse
Affiliation(s)
- Gui-Sheng Li
- School of Pharmacy, Yantai University, 32# Qingquan Road, Laishan District, Yantai 264003, PR China
| | | | | | | | | | | |
Collapse
|
50
|
Guo-Qiu W, Nai-Feng L, Xiao-Bo F, Linxian L, Chen Z, Lixia G, Zhao L. The Level of Connective Tissue Growth Factor in Sera of Patients with Hepatitis B Virus Strongly Correlates with Stage of Hepatic Fibrosis. Viral Immunol 2010; 23:71-8. [PMID: 20121404 DOI: 10.1089/vim.2009.0067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Affiliation(s)
- Wu Guo-Qiu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Liu Nai-Feng
- Institute of Cardiovascular Disease, Zhongda Hospital, Southeast University, Nanjing, China
| | - Fan Xiao-Bo
- Biotechnology Center, Department of Life Science and Biotechnology, China Pharmaceutical University, Nanjing, China
| | - Li Linxian
- Biotechnology Center, Department of Life Science and Biotechnology, China Pharmaceutical University, Nanjing, China
| | - Zhang Chen
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Gou Lixia
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Liu Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|