1
|
Kao C, Ho CH. Time-course RNA sequencing reveals high similarity in mRNAome between hepatic stellate cells activated by agalactosyl IgG and TGF-β1. Funct Integr Genomics 2024; 24:215. [PMID: 39549087 DOI: 10.1007/s10142-024-01502-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Previous studies have demonstrated the clinical relevance of aberrant serum immunoglobulin G (IgG) N-glycomic profiles in liver fibrosis and the pathogenic effects of agalactosyl IgG on activating hepatic stellate cells (HSCs). However, the dynamics of gene expression changes during HSC activation by agalactosyl IgG remain poorly understood. We performed RNA sequencing to analyze the mRNAome of human LX-2 HSCs at multiple time points after treatment with agalactosyl IgG and then compared these results with those obtained after normal IgG and transforming growth factor (TGF)-β1 treatments. Gene expression changes were significantly pronounced on day 5 and subsided by day 11 after HSC activation. A high degree of similarity in gene expression patterns between HSCs treated with agalactosyl IgG and TGF-β1 was observed, of which 1796 and 1785 differentially expressed genes (DEGs) were identified, respectively. Disease ontology analyses revealed that 114 and 105 DEGs in activated HSCs following agalactosyl IgG and TGF-β1 treatments, respectively, were linked to liver cirrhosis, hepatitis, fatty liver disease, hepatitis B, and alcoholic hepatitis, with CCL5 and FAS being the most commonly affected genes. DEGs associated with liver fibrosis or aforementioned liver diseases involved in gene annotation, physiological functions, and signaling pathways regarding secretion of cytokines and chemokines, expression of fibrosis-related growth factors and their receptors, modification of extracellular matrices, and regulation of cell viability in activated HSCs. In conclusion, this study characterized the dynamics of mRNAome and gene networks and identified the liver fibrosis-related DEGs during HSC activation by agalactosyl IgG and TGF-β1.
Collapse
Affiliation(s)
- Chieh Kao
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, No.8, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Cheng-Hsun Ho
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, No.8, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City, 82445, Taiwan.
| |
Collapse
|
2
|
Sharma A, Wang J, Gandhi CR. CD14 is not required for carbon tetrachloride-induced hepatic inflammation and fibrosis with or without lipopolysaccharide challenge. J Cell Physiol 2023; 238:1530-1541. [PMID: 37098757 DOI: 10.1002/jcp.31030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 04/27/2023]
Abstract
Binding of lipopolysaccharide (LPS) to CD14 is required for its cellular effects via TLR4. A role of LPS/TLR4-mediated signaling in activated hepatic stellate cells (aHSCs), the major fibrogenic cells, in liver fibrosis has been reported. We investigated effects of LPS on carbon tetrachloride (CCl4)-induced fibrosis in CD14-knockout (KO) mice in vivo, and culture-activated HSCs in vitro. CCl4 (biweekly; 4 weeks)-treated wild type (WT) and CD14-KO mice were challenged with single LPS administration for 24 h. Liver injury, inflammation and fibrosis were determined. Culture-activated HSCs from WT or CD14-KO mice were stimulated with LPS. Parameters of fibrogenic activity (expression of collagen1a1 [Col1a1], α-smooth muscle actin [αSMA] and TGFβ1) and inflammatory cytokines/chemokines were measured. CCl4 treatment caused similar liver injury and fibrosis in WT and CD14-KO mice. LPS increased liver injury and inflammation similarly in CCl4-treated WT and CD14-KO mice, but downregulated Timp1 and upregulated Mmp13. LPS elicited similar NFκB activation and inflammatory response in WT and CD14-KO aHSCs. LPS similarly downregulated Acta2 (encodes αSMA), Pdgfrb, Col1a1 and Mmp13 expression but did not affect Timp1 expression in WT and CD14-KO aHSCs. LPS did not alter Tgfb1 but increased expression of decorin (Dcn) (inhibitor of TGFβ1) expression in WT and CD14-KO aHSCs. The results indicate that the effects of LPS on HSCs are CD14-independent, and CD14 is not required for hepatic fibrosis. LPS-induced down-modulation of fibrogenic markers in aHSCs is also CD14-independent.
Collapse
Affiliation(s)
- Akanksha Sharma
- Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jiang Wang
- Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Chandrashekhar R Gandhi
- Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
3
|
Rani R, Gandhi CR. Stellate cell in hepatic inflammation and acute injury. J Cell Physiol 2023; 238:1226-1236. [PMID: 37120832 DOI: 10.1002/jcp.31029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 05/02/2023]
Abstract
The perisinusoidal hepatic stellate cells (HSCs) have been investigated extensively for their role as the major fibrogenic cells during chronic liver injury. HSCs also produce numerous cytokines, chemokines, and growth mediators, and express cell adhesion molecules constitutively and in response to stimulants such as endotoxin (lipopolysaccharide). With this property and by interacting with resident and recruited immune and inflammatory cells, HSCs regulate hepatic immune homeostasis, inflammation, and acute injury. Indeed, experiments with HSC-depleted animal models and cocultures have provided evidence for the prominent role of HSCs in the initiation and progression of inflammation and acute liver damage due to various toxic agents. Thus HSCs and/or mediators derived thereof during acute liver damage may be considered as potential therapeutic targets.
Collapse
Affiliation(s)
- Richa Rani
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Research & Development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio, USA
| | - Chandrashekhar R Gandhi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Research & Development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio, USA
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
4
|
Rani R, Sharma A, Wang J, Kumar S, Polaki US, Gandhi CR. Endotoxin-Stimulated Hepatic Stellate Cells Augment Acetaminophen-Induced Hepatocyte Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:518-535. [PMID: 34954210 PMCID: PMC8895430 DOI: 10.1016/j.ajpath.2021.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022]
Abstract
Acetaminophen (APAP)-induced liver injury is influenced by inflammatory Gram-negative bacterial endotoxin [lipopolysaccharide (LPS)], mechanisms of which are not completely understood. Because LPS-stimulated perisinusoidal hepatic stellate cells (HSCs) produce cytokines that affect survival of hepatocytes, this study investigated their role in APAP-induced liver injury. Fed (nonstarved) rats were administered 5 mg/kg LPS or phosphate-buffered saline (PBS) vehicle, followed by 200 mg/kg APAP or PBS an hour later, and euthanized at 6 hours. Control rats received PBS at both time points. Both LPS and APAP caused mild hepatocyte injury (apoptosis), as assessed by histopathology, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining, and caspase-3 activation. The liver injury was augmented in rats administered LPS + APAP, in association with increased nuclear translocation of interferon-regulatory factor-1 (IRF1). In vitro, APAP augmented LPS/HSC-conditioned medium-induced inhibition of DNA and protein synthesis, apoptosis, and nuclear IRF1 in hepatocytes. LPS-stimulated HSCs produced interferon-β (IFN-β), and LPS/HSC + APAP-induced hepatocyte apoptosis was inhibited by anti-IFN-β antibody. Finally, HSC-depleted mice produced significantly lower IFN-β and tumor necrosis factor-α, exhibited less oxidative stress, and were protected from excessive injury due to high APAP dose (600 mg/kg), as well as LPS (5 mg/kg overnight) followed by APAP. In co-culture with or without LPS, HSCs increased expression of proinflammatory cytokines by Kupffer cells. These results suggest that HSCs play a critical role in APAP-induced liver injury without or with LPS preconditioning, and it involves INF-β-IRF1 signaling.
Collapse
Affiliation(s)
- Richa Rani
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Research & development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio
| | - Akanksha Sharma
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Research & development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio
| | - Jiang Wang
- Department of Pathology, University of Cincinnati, Cincinnati, Ohio
| | - Sudhir Kumar
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Research & development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio
| | - Usha S Polaki
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Research & development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio
| | - Chandrashekhar R Gandhi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Research & development, Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio; Department of Surgery, University of Cincinnati, Cincinnati, Ohio; Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
5
|
Sharma A, Verma AK, Kofron M, Kudira R, Miethke A, Wu T, Wang J, Gandhi CR. Lipopolysaccharide Reverses Hepatic Stellate Cell Activation Through Modulation of cMyb, Small Mothers Against Decapentaplegic, and CCAAT/Enhancer-Binding Protein C/EBP Transcription Factors. Hepatology 2020; 72:1800-1818. [PMID: 32064648 PMCID: PMC8009050 DOI: 10.1002/hep.31188] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS During liver injury, quiescent hepatic stellate cells (qHSCs) transdifferentiate into proliferative and fibrogenic activated myofibroblastic phenotype (activated hepatic stellate cell; aHSCs) expressing smooth muscle α-actin (αSMA) and platelet-derived growth factor beta receptor (PDGFβR). Their interactions with gut-derived bacterial lipopolysaccharide (LPS) are implicated in hepatic fibrogenesis. However, LPS can also attenuate fibrogenic characteristics of aHSCs. APPROACH AND RESULTS We examined molecular mechanisms of antifibrogenic effects of LPS on aHSCs in vitro and in vivo. Culture-activated rat HSCs were exposed to 0-100 ng/mL of LPS or its active component, diphosphoryl-lipid A (DPLA), and parameters of fibrosis and inflammatory cytokines/chemokines were determined by qRT-PCR, western, and immunohistochemical analyses. In vivo, HSCs were activated by repeated CCl4 administration to rats every 3 days for 3 or 8 weeks, then challenged with LPS (5 mg/kg; IP). HSCs were isolated 24 hours later, and fibrogenic/inflammatory parameters were analyzed. LPS induced phenotypic changes in aHSCs (rounding, size reduction) and loss of proliferation. LPS down-regulated expression of αSMA, PDGFβR, transforming growth factor beta receptor 1 (TGFβR1), collagen 1α1 (Col1α1), and fibronectin while up-regulating tumor necrosis factor alpha, interleukin-6, and C-X-C motif chemokine ligand 1 expression. LPS did not increase peroxisome proliferation-activated receptor gamma expression or lipid accumulation typical of qHSCs. DPLA elicited the same effects as LPS on aHSCs, indicating specificity, and monophosphoryl lipid A down-regulated fibrogenic markers, but elicited very weak inflammatory response. LPS down-regulated the expression of cMyb, a transcription factor for αSMA, and up-regulated small mother against decapentaplegic (SMAD)7 and CCAAT/enhancer-binding protein (C/EBP)δ, the transcriptional inhibitors of Col1α1 expression. In vivo LPS treatment of aHSCs inhibited their proliferation, down-regulated PDGFβR, αSMA, TGFβR1, Col1α1, and cMyb expression, and increased expression of SMAD7, C/EBPα, and C/EBPδ. CONCLUSIONS In conclusion, LPS induces a unique phenotype in aHSCs associated with down-regulation of key fibrogenic mechanisms and thus may have an important role in limiting fibrosis.
Collapse
Affiliation(s)
- Akanksha Sharma
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatries, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Cincinnati VA Medical Center, Cincinnati, OH
| | - Alok K. Verma
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatries, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Cincinnati VA Medical Center, Cincinnati, OH
| | - Matthew Kofron
- Developmental Biology, Department of Pediatries, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Ramesh Kudira
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatries, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Alexander Miethke
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatries, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Jiang Wang
- Deparment of Pathology, University of Cincinnati, Cincinnati, OH
| | - Chandrashekhar R. Gandhi
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatries, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Cincinnati VA Medical Center, Cincinnati, OH
| |
Collapse
|
6
|
BAY 41-2272 Attenuates CTGF Expression via sGC/cGMP-Independent Pathway in TGFβ1-Activated Hepatic Stellate Cells. Biomedicines 2020; 8:biomedicines8090330. [PMID: 32899801 DOI: 10.3390/biomedicines8090330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Activation of hepatic stellate cells (HSCs) is a critical pathogenic feature of liver fibrosis and cirrhosis. BAY 41-2272 is a canonical non-nitric oxide (NO)-based soluble guanylyl cyclase (sGC) stimulator that triggers cyclic guanosine monophosphate (cGMP) signaling for attenuation of fibrotic disorders; however, the impact of BAY 41-2272 on HSC activation remains ill-defined. Transforming growth factor (TGF)β and its downstream connective tissue growth factor (CTGF or cellular communication network factor 2, CCN2) are critical fibrogenic cytokines for accelerating HSC activation. Here, we identified that BAY 41-2272 significantly inhibited the TGFβ1-induced mRNA and protein expression of CTGF in mouse primary HSCs. Indeed, BAY 41-2272 increased the sGC activity and cGMP levels that were potentiated by two NO donors and inhibited by a specific sGC inhibitor, ODQ. Surprisingly, the inhibitory effects of BAY 41-2272 on CTGF expression were independent of the sGC/cGMP pathway in TGFβ1-activated primary HSCs. BAY 41-2272 selectively restricted the TGFβ1-induced phosphorylation of Akt but not canonical Smad2/3 in primary HSCs. Together, we illustrate a unique framework of BAY 41-2272 for inhibiting TGFβ1-induced CTGF upregulation and HSC activation via a noncanonical Akt-dependent but sGC/cGMP-independent pathway.
Collapse
|
7
|
Gandhi CR. Pro- and Anti-fibrogenic Functions of Gram-Negative Bacterial Lipopolysaccharide in the Liver. Front Med (Lausanne) 2020; 7:130. [PMID: 32373617 PMCID: PMC7186417 DOI: 10.3389/fmed.2020.00130] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
Extensive research performed over several decades has identified cells participating in the initiation and progression of fibrosis, and the numerous underlying inter- and intra-cellular signaling pathways. However, liver fibrosis continues to be a major clinical challenge as the precise targets of treatment are still elusive. Activation of physiologically quiescent perisinusoidal hepatic stellate cells (HSCs) to a myofibroblastic proliferating, contractile and fibrogenic phenotype is a critical event in the pathogenesis of chronic liver disease. Thus, elucidation of the mechanisms of the reversal to quiescence or inhibition of activated HSCs, and/or their elimination via apoptosis has been the focus of intense investigation. Lipopolysaccharide (LPS), a gut-resident Gram-negative bacterial endotoxin, is a powerful pro-inflammatory molecule implicated in hepatic injury, inflammation and fibrosis. In both acute and chronic liver injury, portal venous levels of LPS are elevated due to increased intestinal permeability. LPS, via CD14 and Toll-like receptor 4 (TLR4) and its adapter molecules, stimulates macrophages, neutrophils and several other cell types to produce inflammatory mediators as well as factors that can activate HSCs and stimulate their fibrogenic activity. LPS also stimulates synthesis of pro- and anti-inflammatory cytokines/chemokines, growth mediators and molecules of immune regulation by HSCs. However, LPS was found to arrest proliferation of activated HSCs and to convert them into non-fibrogenic phenotype. Interestingly, LPS can elicit responses in HSCs independent of CD14 and TLR4. Identifying and/or developing non-inflammatory but anti-fibrogenic mimetics of LPS could be relevant for treating liver fibrosis.
Collapse
Affiliation(s)
- Chandrashekhar R Gandhi
- Divisions of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Cincinnati VA Medical Center, Cincinnati, OH, United States
| |
Collapse
|
8
|
Liang Y, Pan Q, Wang R, Ye Z, Li Z, Zeng L, Chen Y, Ma X, Li M, Miao H. Microvesicles Derived from TGF-β1 Stimulated Hepatic Stellate Cells Aggravate Hepatocellular Injury. Stem Cells Dev 2019; 28:1128-1139. [PMID: 31140359 DOI: 10.1089/scd.2019.0032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatic stellate cells (HSCs) are liver-specific cells playing critical roles in liver physiological and pathophysiological processes. Transforming growth factor-β1 (TGF-β1) is an inflammatory cytokine secreted by both hepatocytes and HSCs. We have previously shown that microvesicles (MVs) derived from quiescent HSCs protect hepatocyte functions. In this study, we investigated the effects of MVs released from TGF-β1-stimulated HSCs (HSC-MVs) on xenobiotic-injured hepatocytes. Two hepatocyte cell lines (BRL-3A and HL-7702) were treated with N-acetyl-p-aminophenol or H2O2 to build the injury models. Different concentrations of HSC-MVs were used to coculture with injured hepatocytes. MTT, Hochest33258 staining, and flow cytometry were used to determine their effects on the viability and apoptosis of hepatocytes. Liver injury indicators, alanine aminotransferase (ALT) and aspartate amino transferase (AST), were assessed by enzyme-linked immune sorbent assay kits. The phosphoinositide 3-kinase (PI3K) activator (740Y-P) and extracelluar signal regulated kinase (Erk)1/2 activator (platelet-derived growth factor-BB) were used for pathway analysis. The expression levels of p-PI3K/PI3K, p-Akt/Akt, and activated caspase-3 were measured by western blot. Results showed that (i) HSC-MVs dose dependently impaired the viability of hepatocytes in both injury models, (ii) moreover, HSC-MVs dose dependently increased the apoptosis in those cell models, (iii) HSC-MVs also elevated the levels of ALT and AST in the coculture media, and (iv) these effects were accompanied by a decrease in p-PI3K/PI3K and p-Akt/Akt, which could be partially abolished by 740Y-P. Meanwhile, the proapoptotic effect of HSC-MVs was associated with p-Erk1/2/Erk1/2 downregulation and activated caspase-3 upregulation, and could be inhibited by Erk1/2 activation. Our findings demonstrate that HSC-MVs are involved in inflammatory hepatocytes injury probably through the PI3K/Akt, Erk1/2, and caspase-3 pathways.
Collapse
Affiliation(s)
- Yaolong Liang
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qunwen Pan
- 2Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Rongfeng Wang
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhirong Ye
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zitao Li
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lingdiao Zeng
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yanfang Chen
- 2Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaotang Ma
- 2Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Mingyi Li
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huilai Miao
- 1Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
9
|
Chen Y, Zhao C, Liu X, Wu G, Zhong J, Zhao T, Li J, Lin Y, Zhou Y, Wei Y. Plumbagin ameliorates liver fibrosis via a ROS-mediated NF-кB signaling pathway in vitro and in vivo. Biomed Pharmacother 2019; 116:108923. [PMID: 31154269 DOI: 10.1016/j.biopha.2019.108923] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 01/31/2023] Open
Abstract
AIMS The purpose of this study was to investigate plumbagin (PL) on liver fibrosis in vitro and in vivo and to explore the underlying mechanisms. METHODS Carbon tetrachloride (CCl4) was used to establish a rat liver fibrosis model, primary hepatic stellate cells (HSCs) were isolated from the rat liver, and fibrosis-related indicators were detected. RESULTS The results revealed that PL significantly prevented CCl4-induced liver fibrosis, as evidenced by the attenuation of histopathological changes, the decrease of MDA and the increase of SOD and GSH-P X . In addition, PL downregulated the mRNA levels of NOX4 and procollagen I; the protein expression levels of NOX4 and p-IκB; and the transcriptional activity of NF-κB in liver fibrosis rats. Moreover, PL significantly decreased ROS expression, protein expression of α-SMA and collagen III, and activation of NF-κB and inhibited the nuclear translocation of NF-κB p65 in IL-1β-stimulated HSCs in vitro. CONCLUSION The results of our study indicate that PL can mitigate liver fibrosis in vitro and in vivo, which may be related to the ROS-mediated NF-кB signaling pathway.
Collapse
Affiliation(s)
- Yongxin Chen
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Chuan Zhao
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Xuemei Liu
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Guanyi Wu
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Jing Zhong
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Tiejian Zhao
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Junxuan Li
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Yuning Lin
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Yanping Zhou
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China
| | - Yanfei Wei
- Department of Physiology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China.
| |
Collapse
|
10
|
Kumar S, Rani R, Karns R, Gandhi CR. Augmenter of liver regeneration protein deficiency promotes hepatic steatosis by inducing oxidative stress and microRNA-540 expression. FASEB J 2019; 33:3825-3840. [PMID: 30540918 PMCID: PMC6404588 DOI: 10.1096/fj.201802015r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/29/2018] [Indexed: 12/29/2022]
Abstract
Levels of augmenter of liver regeneration (ALR), a multifunctional protein, are reduced in steatohepatitis. ALR depletion from ALR flox/flox/Alb-Cre [ALR-L-knockout (KO)] mouse causes robust steatosis and apoptosis of hepatocytes, and pericellular fibrosis between 1 and 2 wk postbirth. Steatosis regresses by 4 wk upon reappearance of ALR-expressing hepatocytes. We investigated mechanisms of ALR depletion-induced steatosis. ALR-L-KO mice (1-, 2-, and 4 wk old) and Adeno-Cre-transfected ALR flox/flox hepatocytes were used for in vivo and in vitro studies. ALR depletion from hepatocytes in vivo downregulated peroxisome proliferator-activated receptor (PPAR)-α, carnitine palmitoyl transferase I (CPT1)a, peroxisomal membrane protein 70 (PMP70) (modest down-regulation), and acyl-CoA oxidase 1 (ACOX1). The markedly up-regulated (20X) novel microRNA-540 (miR-540) was identified to target PPARα, PMP70, ACOX1, and CPT1a. ALR depletion from primary hepatocytes increased oxidative stress, miR-540 expression, and steatosis and down-regulated PPARα, ACOX1, PMP70, and CPT1a expression. Anti-miR-540 mitigated ALR depletion-induced steatosis and prevented loss of PPARα, ACOX1, PMP70, and CPT1a expression. Antioxidant N-acetylcysteine and recombinant ALR (rALR) both inhibited ALR depletion-induced miR-540 expression and lipid accumulation in hepatocytes. Finally, treatment of ALR-L-KO mice with rALR between 1 and 2 wk prevented miR-540 expression, and arrested steatosis and fibrosis. We conclude that ALR deficiency-mediated oxidative stress induces generation of miR-540, which promotes steatosis by dysregulating peroxisomal and mitochondrial lipid homeostasis.-Kumar, S., Rani, R., Karns, R., Gandhi, C. R. Augmenter of liver regeneration protein deficiency promotes hepatic steatosis by inducing oxidative stress and microRNA-540 expression.
Collapse
Affiliation(s)
- Sudhir Kumar
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Richa Rani
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Rebekah Karns
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Chandrashekhar R. Gandhi
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
11
|
Rani R, Kumar S, Sharma A, Mohanty SK, Donnelly B, Tiao GM, Gandhi CR. Mechanisms of concanavalin A-induced cytokine synthesis by hepatic stellate cells: Distinct roles of interferon regulatory factor-1 in liver injury. J Biol Chem 2018; 293:18466-18476. [PMID: 30348900 DOI: 10.1074/jbc.ra118.005583] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/15/2018] [Indexed: 12/26/2022] Open
Abstract
Mice depleted of hepatic stellate cells (HSCs) are protected from concanavalin A (ConA)-induced liver injury that is mediated by the activation of interferon regulatory factor 1 (IRF1). The aim of this study was to determine the mechanisms of ConA-mediated signaling and synthesis/release of mediators by HSCs that damage hepatocytes. Primary cultures of wildtype (WT) and IRF1-knockout (KO) HSCs and hepatocytes were used, and ConA-induced liver damage in interferon (IFN)αβ receptor-deficient (IFNαβR-KO) mice was determined. Specific binding of ConA to HSCs induced rapid activation of JAK2 and STAT1. ConA-induced expression of IRF1, IFNβ, tumor necrosis factor α, and CXCL1 was abrogated by selective inhibition of JAK2 and STAT1. Despite activating JAK2/STAT1, ConA failed to stimulate expression of inflammatory cytokines in HSCs from IRF1-KO mice. ConA-conditioned WT-HSC medium caused activation of JNK and caspase 3, and apoptosis of hepatocytes from WT but not from IRF1-KO or IFNαβR-KO mice. Conversely, ConA-conditioned medium of IRF1-KO HSCs failed to cause apoptosis of WT or IRF1-KO hepatocytes. IFNαβR-KO mice were protected from ConA-induced liver damage, and ConA-induced hepatic expression of IRF1 and pro-inflammatory cytokines and chemokines, and infiltration of neutrophils were significantly lower in IFNαβR-KO than in WT mice. These results demonstrate distinct roles of IRF1 in hepatic inflammation (HSCs) and injury (hepatocytes) and can be an important target for intervention in acute liver injury.
Collapse
Affiliation(s)
- Richa Rani
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229.,the Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio 45220 and
| | - Sudhir Kumar
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229.,the Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio 45220 and
| | - Akanksha Sharma
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Sujit K Mohanty
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Bryan Donnelly
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Gregory M Tiao
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Chandrashekhar R Gandhi
- From the Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, .,the Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio 45220 and.,the Department of Surgery, University of Cincinnati, Cincinnati, Ohio 45220
| |
Collapse
|
12
|
Chen Y, Wu Z, Yuan B, Dong Y, Zhang L, Zeng Z. MicroRNA-146a-5p attenuates irradiation-induced and LPS-induced hepatic stellate cell activation and hepatocyte apoptosis through inhibition of TLR4 pathway. Cell Death Dis 2018; 9:22. [PMID: 29348414 PMCID: PMC5833436 DOI: 10.1038/s41419-017-0038-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/14/2022]
Abstract
Elevated toll-like receptor 4 (TLR4) expression is associated with a high risk of radiation-induced liver disease (RILD). MicroRNA (miR)-146a-5p is a key regulator of lipopolysaccharide (LPS)/TLR4 signaling, but its role in modulation of RILD remains unclear. Here, we found that irradiation and LPS stimulation induced TLR4 and miR-146a-5p expression in the human hepatic stellate cell (HSC) line LX2. Ectopic expression of miR-146a-5p in LX2 inhibited irradiation-induced and LPS-induced pro-inflammatory cytokine secretion and cell proliferation, and promoted cell apoptosis by down-regulating the expression levels of TLR4, interleukin-1 receptor associated kinase 1 (IRAK1), tumor necrosis factor receptor associated factor 6 (TRAF6) and phosphorylation of nuclear factor-kappa B. In addition, the culture medium from the irradiated and LPS-stimulated HSCs transfected with miR-146a-5p significantly attenuated apoptosis in irradiated hepatocytes. Overexpression of miR-146a-5p reduced α-smooth muscle actin production in irradiated and LPS-stimulated LX2 cells, which was associated with inhibition of TRAF6-mediated JNK and Smad2 phosphorylation. Knockdown of TRAF6 or IRAK1 mimicked the effects of miR-146a-5p on HSC function. Furthermore, miR-146a-5p treatment alleviated irradiation-induced and endotoxin-induced hepatic inflammatory response and fibrogenesis in mice through inhibition of the TLR4 signaling pathway. Collectively, this study reveals the anti-pro-inflammatory and anti-fibrotic effects of miR-146a-5p on liver injury, and suggests a potential application of miR-146a-5p in the therapeutic prevention of RILD.
Collapse
Affiliation(s)
- Yuhan Chen
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180# Fenglin Road, 200032, Shanghai, China
| | - Zhifeng Wu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180# Fenglin Road, 200032, Shanghai, China
| | - Baoying Yuan
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180# Fenglin Road, 200032, Shanghai, China
| | - Yinying Dong
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180# Fenglin Road, 200032, Shanghai, China
| | - Li Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180# Fenglin Road, 200032, Shanghai, China
| | - Zhaochong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180# Fenglin Road, 200032, Shanghai, China.
| |
Collapse
|
13
|
Vodovotz Y, Simmons RL, Gandhi CR, Barclay D, Jefferson BS, Huang C, Namas R, El-Dehaibi F, Mi Q, Billiar TR, Zamora R. "Thinking" vs. "Talking": Differential Autocrine Inflammatory Networks in Isolated Primary Hepatic Stellate Cells and Hepatocytes under Hypoxic Stress. Front Physiol 2017; 8:1104. [PMID: 29312006 PMCID: PMC5743931 DOI: 10.3389/fphys.2017.01104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/14/2017] [Indexed: 12/23/2022] Open
Abstract
We hypothesized that isolated primary mouse hepatic stellate cells (HSC) and hepatocytes (HC) would elaborate different inflammatory responses to hypoxia with or without reoxygenation. We further hypothesized that intracellular information processing (“thinking”) differs from extracellular information transfer (“talking”) in each of these two liver cell types. Finally, we hypothesized that the complexity of these autocrine responses might only be defined in the absence of other non-parenchymal cells or trafficking leukocytes. Accordingly, we assayed 19 inflammatory mediators in the cell culture media (CCM) and whole cell lysates (WCLs) of HSC and HC during hypoxia with and without reoxygenation. We applied a unique set of statistical and data-driven modeling techniques including Two-Way ANOVA, hierarchical clustering, Principal Component Analysis (PCA) and Network Analysis to define the inflammatory responses of these isolated cells to stress. HSC, under hypoxic and reoxygenation stresses, both expressed and secreted larger quantities of nearly all inflammatory mediators as compared to HC. These differential responses allowed for segregation of HSC from HC by hierarchical clustering. PCA suggested, and network analysis supported, the hypothesis that above a certain threshold of cellular stress, the inflammatory response becomes focused on a limited number of functions in both HSC and HC, but with distinct characteristics in each cell type. Network analysis of separate extracellular and intracellular inflammatory responses, as well as analysis of the combined data, also suggested the presence of more complex inflammatory “talking” (but not “thinking”) networks in HSC than in HC. This combined network analysis also suggested an interplay between intracellular and extracellular mediators in HSC under more conditions than that observed in HC, though both cell types exhibited a qualitatively similar phenotype under hypoxia/reoxygenation. Our results thus suggest that a stepwise series of computational and statistical analyses may help decipher how cells respond to environmental stresses, both within the cell and in its secretory products, even in the absence of cooperation from other cells in the liver.
Collapse
Affiliation(s)
- Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Richard L Simmons
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Chao Huang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rami Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fayten El-Dehaibi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qi Mi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Rani R, Tandon A, Wang J, Kumar S, Gandhi CR. Stellate Cells Orchestrate Concanavalin A-Induced Acute Liver Damage. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2008-2019. [PMID: 28710903 DOI: 10.1016/j.ajpath.2017.05.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/13/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022]
Abstract
Concanavalin A (ConA) causes immune cell-mediated liver damage, but the contribution of resident nonparenchymal cells (NPCs) is also evident. Hepatic stellate cells (HSCs) induce hepatic inflammation and immunological reactions; we therefore investigated their role in ConA-induced liver injury. ConA was administered i.v. to control or HSC-depleted mice; hepatic histopathology and cytokines/chemokines were determined after 6 hours. In vitro, effects of ConA-conditioned HSC medium on hepatocytes were determined. ConA induced inflammation, sinusoidal congestion, and extensive midzonal hepatocyte death in control mice, which were strongly minimized in HSC-depleted mice. CD4 and natural killer T cells and neutrophils were markedly reduced in ConA-treated HSC-depleted mice compared with control mice. The increase in cytokines/chemokines of hepatic injury was much higher in ConA-treated control mice than in HSC-depleted mice. ConA-treated HSCs showed increased expression of interferon-β, tumor necrosis factor-α, and CXCL1, induced oxidative stress in hepatocytes, and caused hepatocyte apoptosis. ConA induced nuclear translocation of interferon-regulatory factor-1 (IRF1) in hepatocytes in vivo, and ConA/HSC induced a similar effect in cultured hepatocytes. IRF1-knockout mice were resistant to ConA-induced liver damage, and anti-interferon β antibody mitigated ConA/HSC-induced injury. In HSC-NPC co-culture, ConA-induced expression of inflammatory cytokines/chemokines was significantly augmented compared with NPCs alone. HSCs play an essential role in ConA-induced liver injury directly via the interferon-β/IRF1 axis, and by modulating properties of NPCs.
Collapse
Affiliation(s)
- Richa Rani
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio; Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ashish Tandon
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Jiang Wang
- Department of Pathology, University of Cincinnati, Cincinnati, Ohio
| | - Sudhir Kumar
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio; Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Chandrashekhar R Gandhi
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio; Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
15
|
Kumar S, Wang J, Shanmukhappa SK, Gandhi CR. Toll-Like Receptor 4-Independent Carbon Tetrachloride-Induced Fibrosis and Lipopolysaccharide-Induced Acute Liver Injury in Mice: Role of Hepatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1356-1367. [PMID: 28412299 PMCID: PMC5455062 DOI: 10.1016/j.ajpath.2017.01.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/30/2017] [Indexed: 12/29/2022]
Abstract
Gram-negative bacterial endotoxin lipopolysaccharide (LPS) is implicated in acute and chronic liver injury; its effects are mediated predominantly via the membrane receptor Toll-like receptor 4 (TLR4). However, TLR4-independent effects of LPS may play important role in hepatic pathophysiology. We investigated carbon tetrachloride (CCl4)-induced fibrosis and LPS-induced acute liver injury in wild-type (WT) and B6.B10ScN-Tlr4lps-del/JthJ [TLR4-knockout (KO)] mice. Effects of LPS on fibrogenic hepatic stellate cells (HSCs) from WT and TLR4-KO mice were assessed in vitro. CCl4 produced similar fibrosis and necroinflammation and increased the mRNA and protein expression of cytokines and chemokines in WT and TLR4-KO mice. However, circulating LPS concentration did not increase in CCl4-treated mice. Interestingly, LPS down-modulated α-smooth muscle actin (activated HSC marker) and collagen 1 in both WT and TLR4-KO HSCs. LPS induced similar activation of NF-κB, and stimulated the expression of cytokines and chemokines in WT and TLR4-KO HSCs. Finally, LPS caused similar inflammation and injury in previously untreated WT and TLR4-KO mice. The results provide evidence of the TLR4/LPS-independent mechanisms of liver fibrosis and also indicate that TLR4 is not entirely critical to LPS-induced acute liver injury. The results further indicate that LPS signaling in activated HSCs might be a mechanism of limiting liver fibrosis.
Collapse
Affiliation(s)
- Sudhir Kumar
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Cincinnati VA Medical Center, Cincinnati, Ohio; Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Shiva Kumar Shanmukhappa
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Chandrashekhar R Gandhi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Cincinnati VA Medical Center, Cincinnati, Ohio; Department of Surgery, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
16
|
Demetris AJ, Bellamy COC, Gandhi CR, Prost S, Nakanuma Y, Stolz DB. Functional Immune Anatomy of the Liver-As an Allograft. Am J Transplant 2016; 16:1653-80. [PMID: 26848550 DOI: 10.1111/ajt.13749] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/26/2016] [Accepted: 01/28/2016] [Indexed: 01/25/2023]
Abstract
The liver is an immunoregulatory organ in which a tolerogenic microenvironment mitigates the relative "strength" of local immune responses. Paradoxically, necro-inflammatory diseases create the need for most liver transplants. Treatment of hepatitis B virus, hepatitis C virus, and acute T cell-mediated rejection have redirected focus on long-term allograft structural integrity. Understanding of insults should enable decades of morbidity-free survival after liver replacement because of these tolerogenic properties. Studies of long-term survivors show low-grade chronic inflammatory, fibrotic, and microvascular lesions, likely related to some combination of environment insults (i.e. abnormal physiology), donor-specific antibodies, and T cell-mediated immunity. The resultant conundrum is familiar in transplantation: adequate immunosuppression produces chronic toxicities, while lightened immunosuppression leads to sensitization, immunological injury, and structural deterioration. The "balance" is more favorable for liver than other solid organ allografts. This occurs because of unique hepatic immune physiology and provides unintended benefits for allografts by modulating various afferent and efferent limbs of allogenic immune responses. This review is intended to provide a better understanding of liver immune microanatomy and physiology and thereby (a) the potential structural consequences of low-level, including allo-antibody-mediated injury; and (b) how liver allografts modulate immune reactions. Special attention is given to the microvasculature and hepatic mononuclear phagocytic system.
Collapse
Affiliation(s)
- A J Demetris
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - C O C Bellamy
- Department of Pathology, University of Edinburgh, Edinburgh, Scotland, UK
| | - C R Gandhi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center and Department of Surgery, University of Cincinnati, Cincinnati, OH
| | - S Prost
- Department of Pathology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Y Nakanuma
- Department of Diagnostic Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | - D B Stolz
- Center for Biologic Imaging, Cell Biology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
17
|
Dangi A, Huang C, Tandon A, Stolz D, Wu T, Gandhi CR. Endotoxin-stimulated Rat Hepatic Stellate Cells Induce Autophagy in Hepatocytes as a Survival Mechanism. J Cell Physiol 2016; 231:94-105. [PMID: 26031389 DOI: 10.1002/jcp.25055] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 05/26/2015] [Indexed: 12/15/2022]
Abstract
Bacterial lipopolysaccharide (LPS)-stimulated hepatic stellate cells (HSCs) produce many cytokines including IFNβ, TNFα, and IL6, strongly inhibit DNA synthesis, but induce apoptosis of a small number of hepatocytes. In vivo administration of LPS (up to 10 mg/mL) causes modest inflammation and weight loss in rats but not mortality. We determined whether LPS-stimulated HSCs instigate mechanisms of hepatocyte survival. Rats received 10 mg/kg LPS (i.p.) and determinations were made at 6 h. In vitro, HSCs were treated with 100 ng/mL LPS till 24 h. The medium was transferred to hepatocytes, and determinations were made at 0-12 h. Controls were HSC-conditioned medium or medium-containing LPS. LPS treatment of rats caused autophagy in hepatocytes, a physiological process for clearance of undesirable material including injured or damaged organelles. This was accompanied by activation of c-Jun NH2 terminal kinase (JNK) and apoptosis of ~4-5% of hepatocytes. In vitro, LPS-conditioned HSC medium (LPS/HSC) induced autophagy in hepatocytes but apoptosis of only ~10% of hepatocytes. While LPS/HSC stimulated activation of JNK (associated with cell death), it also activated NFkB and ERK1/2 (associated with cell survival). LPS-stimulated HSCs produced IFNβ, and LPS/HSC-induced autophagy in hepatocytes and their apoptosis were significantly inhibited by anti-IFNβ antibody. Blockade of autophagy, on the other hand, strongly augmented hepatocyte apoptosis. While LPS-stimulated HSCs cause apoptosis of a subpopulation of hepatocytes by producing IFNβ, they also induce cell survival mechanisms, which may be of critical importance in resistance to liver injury during endotoxemia.
Collapse
Affiliation(s)
- Anil Dangi
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Surgery, University of Cincinnati, and Cincinnati VA Medical Center, Cincinnati, Ohio.,Cincinnati VA Medical Center, Cincinnati, Ohio
| | - Chao Huang
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ashish Tandon
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Surgery, University of Cincinnati, and Cincinnati VA Medical Center, Cincinnati, Ohio
| | - Donna Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Chandrashekhar R Gandhi
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Surgery, University of Cincinnati, and Cincinnati VA Medical Center, Cincinnati, Ohio.,Cincinnati VA Medical Center, Cincinnati, Ohio.,Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
18
|
Sun Y, Xi D, Ding W, Wang F, Zhou H, Ning Q. Soluble FGL2, a novel effector molecule of activated hepatic stellate cells, regulates T-cell function in cirrhotic patients with hepatocellular carcinoma. Hepatol Int 2014; 8:567-575. [PMID: 25298849 PMCID: PMC4182595 DOI: 10.1007/s12072-014-9568-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 07/29/2014] [Indexed: 01/08/2023]
Abstract
PURPOSE To investigate the effects of soluble FGL2 (sFGL2) secreted by hepatic stellate cells (HSCs) on immune suppression in cirrhotic patients with hepatocellular carcinoma (HCC). METHODS Serum sFGL2 levels were examined by ELISA in 40 patients with HCC, liver cirrhosis (LC) or chronic HBV (CHB) infection. A double staining of the immunofluorescence analysis of α-SMA and FGL2 was performed in two cirrhotic liver specimens. The expression of FGL2 in the LX2 cell line was analyzed by immunofluorescence, Western blot and flow cytometry. T-cells purified from HCC patients using magnetic beads were cultured with LX2 cells at different ratios with anti-CD3-stimulating or FGL2-blocking antibodies. The proliferation index (PI) of CD8 + T cells was assessed by flow cytometry, and the secretion of IFN-γ was measured by ELISA. RESULTS sFGL2 levels are significantly higher in patients with HCC or LC compared with those with CHB (p = 0.0039/p = 0.0020). Among HCC patients, those with cirrhosis exhibited significantly higher levels of sFGL2 compared with non-cirrhotic individuals (p = 0.0108). The expressions of FGL2 and α-SMA overlapped in HSCs in liver specimens. FGL2 protein secreted by LX2 cells inhibited T-cell proliferation of HCC patients in a dose-dependent manner in vitro. The PI of CD8 + T cells was significantly enhanced following addition of FGL2 antibody to the culture system (LX2/T-cell ratio of 1:10, p = 0.002). The level of IFN-γ in mixed cultures was inversely correlated with the number of HSCs and was reversed by incubation with FGL2 blocking antibody. CONCLUSION sFGL2 protein is a novel effector molecule of activated HSCs, which suppresses CD8 + T cell proliferation and interferon-γ production, and it subsequently might contribute to immune suppression during fibrosis and tumorigenesis in the liver.
Collapse
Affiliation(s)
- Ying Sun
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, No 1095, Jiefang Avenue, Wuhan, 430030 China
| | - Dong Xi
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, No 1095, Jiefang Avenue, Wuhan, 430030 China
| | - Wen Ding
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, No 1095, Jiefang Avenue, Wuhan, 430030 China
| | - Faxi Wang
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, No 1095, Jiefang Avenue, Wuhan, 430030 China
| | - Haili Zhou
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, No 1095, Jiefang Avenue, Wuhan, 430030 China
| | - Qin Ning
- Department of Infectious Disease, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, No 1095, Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
19
|
Stewart RK, Dangi A, Huang C, Murase N, Kimura S, Stolz DB, Wilson GC, Lentsch AB, Gandhi CR. A novel mouse model of depletion of stellate cells clarifies their role in ischemia/reperfusion- and endotoxin-induced acute liver injury. J Hepatol 2014; 60:298-305. [PMID: 24060854 PMCID: PMC4195246 DOI: 10.1016/j.jhep.2013.09.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 08/05/2013] [Accepted: 09/03/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) that express glial fibrillary acidic protein (GFAP) are located between the sinusoidal endothelial cells and hepatocytes. HSCs are activated during liver injury and cause hepatic fibrosis by producing excessive extracellular matrix. HSCs also produce many growth factors, chemokines and cytokines, and thus may play an important role in acute liver injury. However, this function has not been clarified due to unavailability of a model, in which HSCs are depleted from the normal liver. METHODS We treated mice expressing HSV-thymidine kinase under the GFAP promoter (GFAP-Tg) with 3 consecutive (3 days apart) CCl4 (0.16 μl/g; ip) injections to stimulate HSCs to enter the cell cycle and proliferate. This was followed by 10-day ganciclovir (40 μg/g/day; ip) treatment, which is expected to eliminate actively proliferating HSCs. Mice were then subjected to hepatic ischemia/reperfusion (I/R) or endotoxin treatment. RESULTS CCl4/ganciclovir treatment caused depletion of the majority of HSCs (about 64-72%), while the liver recovered from the initial CCl4-induced injury (confirmed by histology, serum ALT and neutrophil infiltration). The magnitude of hepatic injury due to I/R or endotoxemia (determined by histopathology and serum ALT) was lower in HSC-depleted mice. Their hepatic expression of TNF-α, neutrophil chemoattractant CXCL1 and endothelin-A receptor also was significantly lower than the control mice. CONCLUSIONS HSCs play an important role both in I/R- and endotoxin-induced acute hepatocyte injury, with TNF-α and endothelin-1 as important mediators of these effects.
Collapse
Affiliation(s)
- Rachel K. Stewart
- Thomas E. Starzl Transplantation Institute and Departments of Surgery, University of Pittsburgh, Pittsburgh, PA 15213
| | - Anil Dangi
- Thomas E. Starzl Transplantation Institute and Departments of Surgery, University of Pittsburgh, Pittsburgh, PA 15213,Department of Surgery University of Cincinnati and Cincinnati VA Medical Center, Cincinnati, OH, USA and Cincinnati Veterans Administration, Cincinnati, OH, USA
| | - Chao Huang
- Thomas E. Starzl Transplantation Institute and Departments of Surgery, University of Pittsburgh, Pittsburgh, PA 15213
| | - Noriko Murase
- Thomas E. Starzl Transplantation Institute and Departments of Surgery, University of Pittsburgh, Pittsburgh, PA 15213
| | - Shoko Kimura
- Thomas E. Starzl Transplantation Institute and Departments of Surgery, University of Pittsburgh, Pittsburgh, PA 15213
| | - Donna B. Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gregory C. Wilson
- Department of Surgery University of Cincinnati and Cincinnati VA Medical Center, Cincinnati, OH, USA and Cincinnati Veterans Administration, Cincinnati, OH, USA
| | - Alex B. Lentsch
- Department of Surgery University of Cincinnati and Cincinnati VA Medical Center, Cincinnati, OH, USA and Cincinnati Veterans Administration, Cincinnati, OH, USA
| | - Chandrashekhar R. Gandhi
- Thomas E. Starzl Transplantation Institute and Departments of Surgery, University of Pittsburgh, Pittsburgh, PA 15213,Department of Surgery University of Cincinnati and Cincinnati VA Medical Center, Cincinnati, OH, USA and Cincinnati Veterans Administration, Cincinnati, OH, USA,Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
20
|
The transcriptomic response of rat hepatic stellate cells to endotoxin: implications for hepatic inflammation and immune regulation. PLoS One 2013; 8:e82159. [PMID: 24349206 PMCID: PMC3857241 DOI: 10.1371/journal.pone.0082159] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/26/2013] [Indexed: 12/13/2022] Open
Abstract
With their location in the perisinusoidal space of Disse, hepatic stellate cells (HSCs) communicate with all of the liver cell types both by physical association (cell body as well as cytosolic processes penetrating into sinusoids through the endothelial fenestrations) and by producing several cytokines and chemokines. Bacterial lipopolysaccharide (LPS), circulating levels of which are elevated in liver diseases and transplantation, stimulates HSCs to produce increased amounts of cytokines and chemokines. Although recent research provides strong evidence for the role of HSCs in hepatic inflammation and immune regulation, the number of HSC-elaborated inflammatory and immune regulatory molecules may be much greater then known at the present time. Here we report time-dependent changes in the gene expression profile of inflammatory and immune-regulatory molecules in LPS-stimulated rat HSCs, and their validation by biochemical analyses. LPS strongly up-regulated LPS-response elements (TLR2 and TLR7) but did not affect TLR4 and down-regulated TLR9. LPS also up-regulated genes in the MAPK, NFκB, STAT, SOCS, IRAK and interferon signaling pathways, numerous CC and CXC chemokines and IL17F. Interestingly, LPS modulated genes related to TGFβ and HSC activation in a manner that would limit their activation and fibrogenic activity. The data indicate that LPS-stimulated HSCs become a major cell type in regulating hepatic inflammatory and immunological responses by altering expression of numerous relevant genes, and thus play a prominent role in hepatic pathophysiology including liver diseases and transplantation.
Collapse
|
21
|
Vodovotz Y, Prelich J, Lagoa C, Barclay D, Zamora R, Murase N, Gandhi CR. Augmenter of liver regeneration (ALR) is a novel biomarker of hepatocellular stress/inflammation: in vitro, in vivo and in silico studies. Mol Med 2013; 18:1421-9. [PMID: 23073658 PMCID: PMC3563711 DOI: 10.2119/molmed.2012.00183] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 10/09/2012] [Indexed: 12/15/2022] Open
Abstract
The liver is a central organ involved in inflammatory processes, including the elaboration of acute-phase proteins. Augmenter of liver regeneration (ALR) protein, expressed and secreted by hepatocytes, promotes liver regeneration and maintains viability of hepatocytes. ALR also stimulates secretion of inflammatory cytokines (tumor necrosis factor [TNF]-α and interleukin [IL]-6) and nitric oxide from Kupffer cells. We hypothesized that ALR may be involved in modulating inflammation induced by various stimuli. We found that hepatic ALR levels are elevated at 24 h, before or about the same time as an increase in the mRNA expression of TNF-α and IL-6, after portacaval shunt surgery in rats. Serum ALR also increased, but significantly only on d 4 when pathological changes in the liver become apparent. In rats, serum ALR was elevated after intraperitoneal administration of lipopolysaccharide alone and in a model of gram-negative sepsis. Serum ALR increased before alanine aminotransferase (ALT) in endotoxemia and in the same general time frame as TNF-α and IL-6 in the bacterial sepsis model. Furthermore, mathematical prediction of tissue damage correlated strongly with alterations in serum ALR in a mouse model of hemorrhagic shock. In vitro, monomethyl sulfonate, TNF-α, actinomycin D and lipopolysaccharide all caused increased release of ALR from rat hepatocytes, which preceded the loss of cell viability and/or inhibition of DNA synthesis. ALR may thus serve as a potential diagnostic marker of hepatocellular stress and/or acute inflammatory conditions.
Collapse
Affiliation(s)
- Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John Prelich
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - Claudio Lagoa
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Noriko Murase
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Chandrashekhar R Gandhi
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
22
|
Dangi A, Sumpter TL, Kimura S, Stolz DB, Murase N, Raimondi G, Vodovotz Y, Huang C, Thomson AW, Gandhi CR. Selective expansion of allogeneic regulatory T cells by hepatic stellate cells: role of endotoxin and implications for allograft tolerance. THE JOURNAL OF IMMUNOLOGY 2012; 188:3667-77. [PMID: 22427640 DOI: 10.4049/jimmunol.1102460] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatic stellate cells (HSCs) may play an important role in hepatic immune regulation by producing numerous cytokines/chemokines and expressing Ag-presenting and T cell coregulatory molecules. Due to disruption of the endothelial barrier during cold-ischemic storage and reperfusion of liver grafts, HSCs can interact directly with cells of the immune system. Endotoxin (LPS), levels of which increase in liver diseases and transplantation, stimulates the synthesis of many mediators by HSCs. We hypothesized that LPS-stimulated HSCs might promote hepatic tolerogenicity by influencing naturally occurring immunosuppressive CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Following their portal venous infusion, allogeneic CD4(+) T cells, including Tregs, were found closely associated with HSCs, and this association increased in LPS-treated livers. In vitro, both unstimulated and LPS-stimulated HSCs upregulated Fas (CD95) expression on conventional CD4(+) T cells and induced their apoptosis in a Fas/Fas ligand-dependent manner. By contrast, HSCs induced Treg proliferation, which required cell-cell contact and was MHC class II-dependent. This effect was augmented when HSCs were pretreated with LPS. LPS increased the expression of MHC class II, CD80, and CD86 and stimulated the production of IL-1α, IL-1β, IL-6, IL-10 and TNF-α by HSCs. Interestingly, production of IL-1α, IL-1β, IL-6, and TNF-α was strongly inhibited, but that of IL-10 enhanced in LPS-pretreated HSC/Treg cocultures. Adoptively transferred allogeneic HSCs migrated to the secondary lymphoid tissues and induced Treg expansion in lymph nodes. These data implicate endotoxin-stimulated HSCs as important immune regulators in liver transplantation by inducing selective expansion of tolerance-promoting Tregs and reducing inflammation and alloimmunity.
Collapse
Affiliation(s)
- Anil Dangi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
In the steady state, hepatic antigen (Ag)-presenting cells (APC) generally dampen systemic inflammatory responses to gut-derived Ags. Our studies focus on the role of specific liver APC populations, both non-parenchymal cells (dendritic cells [DC], Kupffer cells, and hepatic stellate cells [HSC]) and parenchymal cells, in the molecular regulation of tissue damage (ischemia and reperfusion [I/R] injury) and immunity following liver transplantation. We focus on factors that either promote or overwhelm the natural tendency of the liver to suppress inflammatory/immune responses. We are also examining molecular mechanisms that regulate liver DC maturation and function and that determine their role in the control of allogeneic T-cell function and the fate of the transplanted liver. Our studies are also aimed at elucidating mechanisms by which HSC regulate DC and T-cell function. These investigations may provide new targets for therapeutic intervention in liver inflammation.
Collapse
|
24
|
Inhibition of T-cell responses by intratumoral hepatic stellate cells contribute to migration and invasion of hepatocellular carcinoma. Clin Exp Metastasis 2011; 28:661-74. [PMID: 21717117 DOI: 10.1007/s10585-011-9399-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 06/13/2011] [Indexed: 02/07/2023]
Abstract
The stroma of hepatocellular carcinoma (HCC) is markedly infiltrated with activated hepatic stellate cells (HSCs), and associated invasion and metastasis of HCC. However, little is known of the role of HSCs in immune responses in HCC. The Buffalo rat HCC model was established. Quiescent HSCs (qHSCs) and intratumoral HSCs (tHSCs) were isolated. Surface molecules of tHSC were detected by flow cytometry, and gene expression was analyzed by fluorescence quantitative RT-PCR. T cell proliferation was monitored by [(3)H]-thymidine ((3)H-TdR) incorporation into DNA, and cytotoxic activity was assessed by measuring the release of (51)Cr. The level of cytokine expression by T cells was measured by enzyme-linked immunosorbent assay. T cell apoptosis was detected by double-stained terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and anti-CD3 antibodies. The migration and invasion of HCC was observed by transwell experiments. tHSCs express low levels of major histocompatibility complex (MHC) class I, MHC class II, and costimulatory molecules, and produce varying levels of cytokines. Addition of the tHSCs suppressed thymidine uptake by T cells that were stimulated by alloantigens or by anti-CD3-mediated T-cell receptor ligation. The tHSC-induced T-cell hyporesponsiveness was associated with enhanced T-cell apoptosis, and contributed to the migration and invasion of hepatoma cell. tHSCs was associated with markedly enhanced expression of B7-H1. Blockade of B7-H1/PD-1 ligation significantly reduced HSC immunomodulatory activity, and hepatoma cell migration and invasion. tHSCs can induce T cell apoptosis, suggesting an important role for B7-H1. The interactions between tHSCs and T cells may contribute to hepatic immune tolerance and invasion and migration of HCC.
Collapse
|
25
|
Jameel NM, Thirunavukkarasu C, Murase N, Cascio M, Prelich J, Yang S, Harvey SAK, Gandhi CR. Constitutive release of powerful antioxidant-scavenging activity by hepatic stellate cells: protection of hepatocytes from ischemia/reperfusion injury. Liver Transpl 2010; 16:1400-9. [PMID: 21117250 DOI: 10.1002/lt.22172] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Within the liver, reactive oxygen species produced by infiltrating blood cells and Kupffer cells (resident macrophages) can injure hepatocytes. We hypothesized that hepatocyte survival is influenced by the relatively small juxtaposed population of hepatic stellate cells (HSCs). We used cultures of primary rat hepatocytes as targets for superoxide-induced damage, which was determined by crystal violet assay and lactate dehydrogenase release. An HSC-conditioned medium prevented the superoxide-induced death of hepatocytes, and the protective factor released by HSCs was a protein or proteins (apparent molecular weight > 100 kDa) resistant to heat (70°C) and pH (4.5-8.5). The protein or proteins were partially purified on DE52 cellulose, and the active fraction contained no detectable levels of superoxide dismutase: after separation by Sephadex G-100 gel filtration, the antioxidant activity could be reconstituted by the combination of 2 protein peaks, and this reconstituted activity was protective both in vitro and against liver ischemia/reperfusion injury in intact rats. Mass spectrometry proteomic studies confirmed that this activity could not be attributed to any previously identified antioxidant protein. Thus, HSCs protect hepatocytes against oxidative damage through the production of a novel protein, the further purification of which may lead to the isolation of a powerful oxygen radical scavenger with clinical applications.
Collapse
Affiliation(s)
- Noor Mohamed Jameel
- Thomas E. Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Gandhi CR, Murase N, Starzl TE. Cholera toxin-sensitive GTP-binding protein-coupled activation of augmenter of liver regeneration (ALR) receptor and its function in rat kupffer cells. J Cell Physiol 2010; 222:365-73. [PMID: 19859909 PMCID: PMC3034370 DOI: 10.1002/jcp.21957] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mitogenic effect of augmenter of liver regeneration (ALR), a protein produced and released by hepatocytes, on hepatocytes in vivo but not in vitro suggests that the effect is mediated by nonparenchymal cells. Since mediators produced by Kupffer cells are implicated in hepatic regeneration, we investigated receptor for ALR and its functions in rat Kupffer cells. Kupffer cells were isolated from rat liver by enzymatic digestion and centrifugal elutriation. Radioligand ([(125)I] ALR) receptor binding, ALR-induced GTP/G-protein association, and nitric oxide (NO), tumor necrosis factor (TNF)-alpha, and interleukin-6 (IL-6) synthesis were determined. High-affinity receptor for ALR, belonging to the G-protein family, with K(d) of 1.25 +/- 0.18 nM and B(max) of 0.26 +/- 0.02 fmol/microg DNA was identified. ALR stimulated NO, TNF-alpha, and IL-6 synthesis via cholera toxin-sensitive G-protein, as well as p38-MAPK activity and nuclear translocation of NFkappaB. While inhibitor of NFkappaB (MG132) inhibited ALR-induced NO synthesis, MG132 and p38-MAPK inhibitor (SB203580) abrogated ALR-induced TNF-alpha and IL-6 synthesis. ALR also prevented the release of mediator(s) from Kupffer cells that cause inhibition of DNA synthesis in hepatocytes. Administration of ALR to 40% partially hepatectomized rats increased expression of TNF-alpha, IL-6, and inducible nitric oxide synthase (iNOS) and caused augmentation of hepatic regeneration. These results demonstrate specific G-protein coupled binding of ALR and its function in Kupffer cells and suggest that mediators produced by ALR-stimulated Kupffer cells may elicit physiologically important effects on hepatocytes.
Collapse
Affiliation(s)
- Chandrashekhar R. Gandhi
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pathology, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Noriko Murase
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas E. Starzl
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Chen Y, Wang CP, Lu YY, Zhou L, Su SH, Jia HJ, Feng YY, Yang YP. Hepatic stellate cells may be potential effectors of platelet activating factor induced portal hypertension. World J Gastroenterol 2008; 14:218-23. [PMID: 18186558 PMCID: PMC2675117 DOI: 10.3748/wjg.14.218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine platelet activating factor (PAF) receptor expression in cirrhotic hepatic stellate cells.
METHODS: Hepatic stellate cells, isolated from the livers of control and CCl4-induced cirrhotic rats, were placed in serum-free medium after overnight culture. We determined the PAF receptor in hepatic stellate cells by saturation binding technique and semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR), and the effects of PAF and its antagonist BN52021 on prostaglandin E2 (PGE2) release by stellate cells.
RESULTS: Scatchard analysis indicated the presence of PAF receptor with dissociation constant (Kd) of 4.66 nmol/L and maximum binding capacity (Bmax) of 24.65 fmol/&mgr;g in cirrhotic stellate cells. Compared with the control, the maximum PAF binding capacity increased significantly (Bmax: 24.65 ± 1.96 fmol/&mgr;g. DNA, R = 0.982 vs 5.74 ± 1.55 fmol/&mgr;g. DNA, R = 0.93; P < 0.01), whereas receptor affinity had no significant difference (Kd of 4.66 ± 0.33 nmol/L for the cirrhosis and 3.51 ± 0.26 nmol/L for the control; P > 0.05). Consistent with the receptor binding data, the mRNA expression of PAF receptor was increased significantly in cirrhotic stellate cells. PAF in a concentration-dependent manner induced PGE2 synthesis in cirrhotic hepatic stellate cells, but the effects were blocked significantly by BN52021.
CONCLUSION: Cirrhosis sensitizes hepatic stellate cells to PAF by elevating its receptor level and hepatic stellate cells maybe potential effectors of PAF induced portal hypertension.
Collapse
|
29
|
Zeng G, Awan F, Otruba W, Muller P, Apte U, Tan X, Gandhi C, Demetris AJ, Monga SPS. Wnt'er in liver: expression of Wnt and frizzled genes in mouse. Hepatology 2007; 45:195-204. [PMID: 17187422 DOI: 10.1002/hep.21473] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED The Wnt signaling pathway is essential for a wide array of developmental and physiological processes. Wnts are extracellular ligands that bind to frizzled (Fz) receptors at the membrane, canonically inducing beta-catenin nuclear translocation and activation. Although beta-catenin has been shown to be critical in liver biology, the expression of the 19 Wnt and 10 Fz genes in liver remains undetermined. We report comprehensive analysis of Wnt and Fz expression in whole liver as well as individual cell types: freshly isolated and plated hepatocytes, biliary epithelial cells, normal and activated stellate and Kupffer cells, and sinusoidal endothelial cells (SECs). Oligonucleotides for the 19 Wnt, 10 frizzled receptors genes, and secreted Frizzled-related protein-1 (sFRP or Fzb) were synthesized based on the available sequences. A total of 11 Wnts and 8 Fz genes and Fzb were expressed in normal liver. Although only 6 Wnt and 5 Fz genes were expressed in freshly isolated hepatocytes, 8 Wnt genes, 7 Fz genes, and Fzb were expressed in plated hepatocytes. Although 12 Wnt and 7 Fz genes were expressed in biliary tree, additional Fz9 and Fzb were only expressed in cultured biliary epithelial cells. The same 14 Wnt and 7 Fz genes were expressed in both activated and normal stellate and Kupffer cells; only Fzb was expressed in their activated state. Also, 11 Wnt, seven Fz, and Fzb genes were expressed in SECs. CONCLUSION These data indicate that most Wnt and frizzled genes are expressed in the liver and might be playing important roles in liver pathobiology via canonical and noncanonical pathways.
Collapse
Affiliation(s)
- Gang Zeng
- Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
BACKGROUND/AIMS This study determined the roles of NAD(P)H oxidase, which generates reactive oxygen species (ROS), and of inducible nitric oxide synthase (iNOS), which generates nitric oxide (NO) on the development of hepatic fibrosis in mice. METHODS Hepatic fibrosis was produced by carbon tetrachloride administered for 12 weeks in wild-type (WT) mice and in mice with knockout of either the gp91phox subunit of the NAD(P)H complex (gp91phox-/-) or of iNOS (iNOS(-/-)). RESULTS Liver fibrosis and hydroxyproline after carbon tetrachloride was lower in gp91phox-/- and in iNOS(-/-) mice than in WT mice. The increase in alpha2(I) collagen mRNA was absent in the gp91phox-/- but not in the iNOS(-/-) mice. Transformation growth factor beta (TGF-beta) mRNA was increased more in the gp91phox-/- than in the WT mice, while in the iNOS(-/-) mice there was no increase in TGF-beta mRNA. 3-Nitrotyrosine was similarly increased by carbon tetrachloride in gp91phox-/- and WT mice, while there was no increase in the iNOS(-/-) mice. CONCLUSIONS Deficiencies in NAD(P)H oxidase and in iNOS separately reduce, but do not eliminate carbon tetrachloride-induced liver fibrosis. Likely causes for this inhibitory effects are decreases in the production of ROS in NAD(P)H deficiency and of peroxinitrite radicals in iNOS deficiency.
Collapse
Affiliation(s)
- Gennadiy Novitskiy
- Department of Medicine, The Johns Hopkins University School of Medicine, The Johns Hopkins University, Baltimore 21205-2195, MD, USA
| | | | | | | |
Collapse
|
31
|
Thirunavukkarasu C, Watkins SC, Gandhi CR. Mechanisms of endotoxin-induced NO, IL-6, and TNF-alpha production in activated rat hepatic stellate cells: role of p38 MAPK. Hepatology 2006; 44:389-98. [PMID: 16871588 DOI: 10.1002/hep.21254] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Compelling experimental evidence indicates that the interactions between endotoxin and hepatic stellate cells (HSCs) can play a significant role in the pathogenesis of liver disease. Endotoxin-induced release of a multifunctional mediator NO (via inducible NO synthase) and the proinflammatory cytokines tumor necrosis factor alpha (TNF-alpha) and interleukin (IL)-6 by HSCs could be an important mechanism of pathological changes in the liver. However, the signaling mechanisms of these effects are poorly understood. In this study, we found that endotoxin causes activation of mitogen-activated protein kinases (MAPKs) (extracellular signal-regulated protein kinase [ERK] 1 and 2, p38, and c-Jun NH2-terminal kinase [JNK]) and nuclear factor kappaB (NF-kappaB) and production of H(2)O(2) in culture-activated HSCs. However, only p38 and NF-kappaB were found to be responsible for the synthesis of NO, IL-6, and TNF-alpha. Exogenous H(2)O(2) caused modest stimulation of TNF-alpha synthesis, did not affect the synthesis of NO or IL-6, and did not activate NF-kappaB or MAPKs. Inhibition of p38 and NF-kappaB activation by SB203580 and pyrrolidine dithiocarbamate, respectively, blocked endotoxin-induced H(2)O(2), NO, TNF-alpha, and IL-6 synthesis. Inhibition of ERK1/2 and JNK phosphorylation did not alter these effects of endotoxin. Whereas SB203580 inhibited endotoxin-induced NF-kappaB activation, pyrrolidine dithiocarbamate did not affect p38 phosphorylation in endotoxin-stimulated cells. In conclusion, endotoxin-induced synthesis of NO, TNF-alpha, and IL-6 in HSCs is mediated by p38 and NF-kappaB, with involvement of H(2)O(2) in TNF-alpha production.
Collapse
Affiliation(s)
- Chinnasamy Thirunavukkarasu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
32
|
Thirunavukkarasu C, Uemura T, Wang LF, Watkins SC, Gandhi CR. Normal rat hepatic stellate cells respond to endotoxin in LBP-independent manner to produce inhibitor(s) of DNA synthesis in hepatocytes. J Cell Physiol 2005; 204:654-65. [PMID: 15828022 DOI: 10.1002/jcp.20366] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Endotoxin is implicated in the pathology of acute liver failure. The mechanisms of its actions on quiescent hepatic stellate cells (qHSCs) and their implications in hepatocyte injury are incompletely understood. We investigated effects of endotoxin (bacterial lipopolysaccharide; LPS) on qHSCs and subsequently on hepatocytes. After overnight culture following their isolation, qHSCs were incubated with or without endotoxin for 24 h. The cells and the culture supernatant were analyzed for cytokines and nitric oxide (NO) synthesis. The effects of qHSC-conditioned media on hepatocytes were then determined. LPS increased inducible NO synthase expression, stimulated NO synthesis, and inhibited DNA synthesis in qHSCs. qHSC-conditioned medium inhibited DNA synthesis in hepatocytes without affecting NO synthesis, while LPS (1-1,000 ng/ml)-conditioned qHSC medium stimulated NO synthesis and caused further inhibition of DNA synthesis and apoptosis. These effects of LPS were more pronounced when qHSCs were incubated with serum, but not with LPS-binding protein (LBP) although CD14 (a receptor for LPS-LBP complex) was found in qHSCs. LPS stimulated the synthesis of TNF-alpha, interleukin (IL)-6, and IL-1beta but not of TGF-beta in qHSCs. Individually or together, L-N(G)-monomethylarginine and antibodies to IL-1beta, IL-6, and TNF-alpha only partly reversed qHSC + LPS-conditioned medium-induced inhibition of DNA synthesis in hepatocytes. These results suggest that the effects of LPS on qHSCs are novel, occurring without the aid of LBP/CD14. They also indicate that other factors, in addition to NO, TGF-beta, TNF-alpha, IL-1beta, and IL-6 are involved in the mechanisms of the growth inhibitory effects of qHSCs on hepatocytes.
Collapse
Affiliation(s)
- Chinnasamy Thirunavukkarasu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | | | |
Collapse
|
33
|
Sturm E, Havinga R, Baller JFW, Wolters H, van Rooijen N, Kamps JAAM, Verkade HJ, Karpen SJ, Kuipers F. Kupffer cell depletion with liposomal clodronate prevents suppression of Ntcp expression in endotoxin-treated rats. J Hepatol 2005; 42:102-9. [PMID: 15629514 DOI: 10.1016/j.jhep.2004.09.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2004] [Revised: 08/26/2004] [Accepted: 09/17/2004] [Indexed: 01/02/2023]
Abstract
BACKGROUND/AIMS In sepsis-associated cholestasis, expression of many genes involved in bile acid transport, including Ntcp, is suppressed by cytokines. Kupffer cells (KC) are an important source of cytokines in sepsis. To assess the consequences of KC depletion on hepatic Ntcp expression in endotoxemic rats. METHODS Sprague-Dawley rats received liposomal clodronate (CLO) or vehicle (PBS) to deplete KC prior to lipopolysaccharide (LPS) exposure. Plasma and liver samples were taken 1 and 16 h after LPS exposure. RESULTS Complete CLO-depletion of KC by was demonstrated by immunohistochemistry. Hepatic gene expression of IL-1beta and TNFalpha as well as TNFalpha plasma levels in CLO/LPS-injected animals were significantly reduced to a mean of 41, 36 and 23% of controls injected with LPS only. Ntcp RNA- and protein expression was significantly higher whereas plasma bile salt concentration was lower in CLO/LPS animals vs. animals injected with LPS only. Binding activity of transcription factors RXR:RAR and HNF1alpha was decreased in LPS only controls but preserved in CLO/LPS treated animals. CONCLUSIONS Clodronate-depletion of KC blocks cytokine-mediated Ntcp suppression upon endotoxin exposure. KC may represent pharmacological targets for treatment of sepsis-associated cholestasis.
Collapse
Affiliation(s)
- Ekkehard Sturm
- Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yu MC, Chen CH, Liang X, Wang L, Gandhi CR, Fung JJ, Lu L, Qian S. Inhibition of T-cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology 2004; 40:1312-21. [PMID: 15565659 DOI: 10.1002/hep.20488] [Citation(s) in RCA: 248] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the injured liver, hepatic stellate cells (HSCs) secrete many different cytokines, recruit lymphocytes, and thus participate actively in the pathogenesis of liver disease. Little is known of the role of HSCs in immune responses. In this study, HSCs isolated from C57BL/10 (H2b) mice were found to express scant key surface molecules in the quiescent stage. Activated HSCs express major histocompatibility complex class I, costimulatory molecules, and produce a variety of cytokines. Stimulation by interferon gamma (IFN-gamma) or activated T cells enhanced expression of these molecules. Interestingly, addition of the activated (but not quiescent) HSCs suppressed thymidine uptake by T cells that were stimulated by alloantigens or by anti-CD3-mediated T-cell receptor ligation in a dose-dependent manner. High cytokine production by the T cells suggests that the inhibition was probably not a result of suppression of their activation. T-cell division was also found to be normal in a CFSE dilution assay. The HSC-induced T-cell hyporesponsiveness was associated with enhanced T-cell apoptosis. Activation of HSCs was associated with markedly enhanced expression of B7-H1. Blockade of B7-H1/PD-1 ligation significantly reduced HSC immunomodulatory activity, suggesting an important role of B7-H1. In conclusion, the bidirectional interactions between HSCs and immune cells may contribute to hepatic immune tolerance.
Collapse
Affiliation(s)
- Ming-Chin Yu
- Thomas E. Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Thirunavukkarasu C, Watkins S, Harvey SAK, Gandhi CR. Superoxide-induced apoptosis of activated rat hepatic stellate cells. J Hepatol 2004; 41:567-75. [PMID: 15464236 DOI: 10.1016/j.jhep.2004.06.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Revised: 05/19/2004] [Accepted: 06/02/2004] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIMS During liver injury, reactive oxygen species (ROS) are produced by the resident macrophages (Kupffer cells) and infiltrating blood cells such as neutrophils. ROS cause transformation of desmin-positive quiescent hepatic stellate cells (HSCs) into the proliferating activated phenotype that expresses alpha-smooth muscle actin (alpha-SMA). The highly fibrogenic and contractile activated HSCs (aHSCs) produce various cytokines and growth factors, and play important role in the pathophysiology of chronic liver disease. However, apoptotic aHSCs are also observed during active fibrogenesis in the injured liver. Therefore, we investigated the mechanisms of apoptosis of aHSCs in relation to ROS. METHODS HSCs, isolated from normal rat liver, were activated in culture and effects of superoxide were determined between subcultures 3 and 5. RESULTS Treatment with superoxide caused apoptosis of aHSCs as determined by flow cytometry, TUNEL assay and DNA laddering analysis. The mechanisms of superoxide-induced apoptosis involved release of cytochrome c, increased Bax expression, increased caspase-3 activity, and hydrolysis of polyADP-ribose polymerase. Superoxide also increased the expression of antiapoptotic Bcl-xL and nuclear translocation of NFkappaB. Caspase-3 inhibitor (DEVD-fmk) and antioxidants (N-acetylcysteine, vitamin E and superoxide dismutase) inhibited superoxide-induced apoptosis. CONCLUSIONS Superoxide-induced apoptosis of aHSCs may be a novel mechanism of limiting chronic fibrotic liver injury.
Collapse
Affiliation(s)
- Chinnasamy Thirunavukkarasu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, E-1542 BST, 200 Lothrop street, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
36
|
|
37
|
Yang Y, Nemoto EM, Harvey SAK, Subbotin VM, Gandhi CR. Increased hepatic platelet activating factor (PAF) and PAF receptors in carbon tetrachloride induced liver cirrhosis. Gut 2004; 53:877-83. [PMID: 15138217 PMCID: PMC1774060 DOI: 10.1136/gut.2003.024893] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2003] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS The liver is a major site for the synthesis and actions of platelet activating factor (PAF), a potent hepatic vasoconstrictor and systemic vasodilator. As PAF is implicated in portal hypertension and hyperdynamic circulation associated with liver cirrhosis, we characterised changes in the hepatic PAF system in experimental cirrhosis. METHODS In rats made cirrhotic by carbon tetrachloride (CCl(4)) administration for eight weeks, we determined hepatic levels of PAF and its cognate receptor, and the effects of PAF and PAF antagonist (BN52021) on portal and arterial pressure. RESULTS Compared with control rats, cirrhotic rats had higher hepatic PAF levels, higher apparent hepatic efflux of PAF, and higher PAF levels in arterial blood (p<0.01, p<0.01, p<0.05, respectively). Relative to controls, cirrhotic livers had elevated hepatic PAF receptors (by mRNA and protein levels and [(3)H]PAF binding), higher (p<0.01) baseline hepatic portal pressure, and an augmented (p = 0.03) portal pressure response to PAF infusion (1 microg/kg). Portal infusion of BN52021 (5 mg/kg) showed that elevated endogenous PAF was responsible for 23% of the cirrhotic portal pressure increase but made no contribution to systemic hypotension. Finally, increased PAF receptor density was observed in the contractile perisinusoidal stellate cells isolated from cirrhotic livers relative to those from control livers. CONCLUSIONS In cirrhosis, increased hepatic release of PAF elevates systemic PAF; in combination with upregulated hepatic PAF receptors in stellate cells, this contributes to portal hypertension.
Collapse
Affiliation(s)
- Y Yang
- Thomas E Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
38
|
Chi X, Anselmi K, Watkins S, Gandhi CR. Prevention of cultured rat stellate cell transformation and endothelin-B receptor upregulation by retinoic acid. Br J Pharmacol 2003; 139:765-74. [PMID: 12813000 PMCID: PMC1573899 DOI: 10.1038/sj.bjp.0705303] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
1 Physiologically, perisinusoidal hepatic stellate cells (HSC) are quiescent and store retinoids. During liver injury and in cell culture, HSC transform into proliferating myofibroblast-like cells that express alpha-smooth muscle actin (alpha-sma) and produce excessive amounts of extracellular matrix. During transformation (also known as activation), HSC are depleted of the retinoid stores, and their expression of the endothelin-1 (ET-1) system is increased. ET-1 causes contraction of transformed HSC and is implicated in their proliferation and fibrogenic activity. In order to understand the association between retinoids, ET-1 and the activation of HSC, we investigated the effect of 13-cis-retinoic acid on the transformation of cultured HSC and the expression of ET-1 system. 2 HSC derived from normal rat liver were maintained for 10-12 days in a medium supplemented with 5% serum and containing 2.5 micro M retinoic acid without or with 50 nM ET-1 (ETA+ETB agonist) or sarafotoxin S6c (ETB agonist). In another set of experiments, cells treated for 10-12 days with vehicle (ethanol) or retinoic acid were challenged with ET-1 or sarafotoxin S6c, and various determinations were made at 24 h. 3 Retinoic acid inhibited transformation and proliferation of HSC as assessed by morphological characteristics, expression of alpha-sma, bromodeoxyuridine incorporation and cell count. Retinoic acid also prevented upregulation of ETB receptors without affecting ET-1 or ETA expression. Total protein synthesis ([(3)H]leucine incorporation), collagen alpha types I mRNA expression and collagen synthesis ([(3)H]proline incorporation) were lower in retinoic acid-treated cells. Although ET-1-treated cells were morphologically similar to the control cells, their expression of alpha-smooth muscle actin was significantly inhibited. The presence of retinoic acid in the medium during treatment with ET-1 caused further reduction in the expression of alpha-smooth muscle actin. ET-1 and sarafotoxin S6c stimulated total protein synthesis in vehicle- and retinoic acid-treated cells, but collagen synthesis only in the latter. 4 These results showing prevention of HSC activation and negative regulation of ETB receptor expression in them by retinoic acid may have important pathophysiologic implications.
Collapse
Affiliation(s)
- Xuedong Chi
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, E-1540 BST, 200 Lothrop Street, Pittsburgh, PA 15213, U.S.A
| | - Kristin Anselmi
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, E-1540 BST, 200 Lothrop Street, Pittsburgh, PA 15213, U.S.A
| | - Simon Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Chandrashekhar R Gandhi
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, E-1540 BST, 200 Lothrop Street, Pittsburgh, PA 15213, U.S.A
- Department of Pathology, Veterans Administration Medical Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Author for correspondence:
| |
Collapse
|