1
|
Mounika N, Mungase SB, Verma S, Kaur S, Deka UJ, Ghosh TS, Adela R. Inflammatory Protein Signatures as Predictive Disease-Specific Markers for Non-Alcoholic Steatohepatitis (NASH). Inflammation 2025; 48:25-41. [PMID: 38676759 DOI: 10.1007/s10753-024-02035-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic disease worldwide, consisting of a broad spectrum of diseases such as simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. Hepatic inflammation plays a key role in the pathophysiology of NAFLD. Inflammatory mediators such as cytokines and chemokines are considered as contributing factors to NAFLD development and progression. In the present study, we aimed to investigate the inflammatory protein signatures as predictive disease-specific markers for non-alcoholic fatty liver disease (NAFLD). This cross-sectional study included healthy control (n = 64), NAFL (n = 109), and NASH (n = 60) human subjects. Serum concentrations of various cytokines and chemokines were evaluated using sensitive multiplex assays. We used principal component analysis (PCoA) to reveal distinct differences in the levels of cytokines and chemokines between each of the study groups. Further, a random forest classification model was developed to identify the panel of markers that could predict diseases. The protein-protein network analysis was performed to determine the various signaling pathways associated with the disease-specific panel of markers. Serum concentrations of TNF-α, IL-1β, IL-1ra, G-CSF, PDGF-BB, MCP-1, MIP-1a, MIP-1b, RANTES, eotaxin, IL-8 and IP-10 were significantly increased in NASH group as compared to control group. Furthermore, serum concentrations of IL-9 and IL-13 were significantly lower in the NASH group, whereas IL-2 levels were significantly decreased in the NAFL group when compared to the control group. PCoA results demonstrated statistically significant differences in cytokines and chemokines between each of the study groups (PERMANOVA p = 0.001; R2 = 0.102). RANTES, IL-1ra, MIP-1b, IL-2, and G-CSF could differentiate the NAFL group from the controls; G-CSF, IL-1ra, TNF-α, RANTES, and IL-9 could differentiate the NASH group from the controls; and G-CSF, IL-9, IL-13, eotaxin, and TNF- α could differentiate the NASH group from the NAFL group. Our protein-protein network revealed that these markers are involved in cytokine-cytokine receptor interaction, Th1 and Th2 cell differentiation, TNF, chemokine, JAK/STAT, P13K/Akt, TLR, NOD-like receptor, NF-kB, and adipocytokine signaling pathways which might be responsible for disease pathogenesis. Our study findings revealed a set of distinct cytokine and chemokine markers and they might be considered as biomarkers in distinguishing NASH from NAFL. Future multicentre studies with larger sample size are recommended to determine the potential utility of these panels of markers.
Collapse
Affiliation(s)
- Nadella Mounika
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India
| | - Suraj Bhausaheb Mungase
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India
| | - Shivangi Verma
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), Okhla Phase III, New Delhi, 110020, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver & Biliary Science (ILBS), New Delhi-110 070, Vasant Kunj, India
| | - Utpal Jyoti Deka
- Department of Gastroenterology, Downtown Hospital, GS Road, Bormotoria, Guwahati, Assam-781006, India
| | - Tarini Shankar Ghosh
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), Okhla Phase III, New Delhi, 110020, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur (Halugurisuk), Changsari, Kamrup, Assam-781101, India.
| |
Collapse
|
2
|
Virseda-Berdices A, Martín-Escolano R, Berenguer J, González-García J, Brochado-Kith O, Rojo D, Díez C, Hontañon V, Pérez-Latorre L, Ibañez-Samaniego L, Llop-Herrera E, Olveira A, Fernández-Rodríguez A, Barbas C, Resino S, Jiménez-Sousa MÁ. Metabolomic Changes Associated With the Change in HVPG After DAAs Therapy in HCV Cirrhotic Patients. Liver Int 2025; 45:e16204. [PMID: 39708286 DOI: 10.1111/liv.16204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND AND AIMS In response to direct-acting antivirals (DAAs) therapy, patients who experience a decrease in hepatic venous pressure gradient (HVPG) considerably reduce liver complications and have increased survival. This study aimed to assess the metabolomic changes associated with the changes in HVPG from the start of DAA therapy until 48 weeks after effective DAA therapy in patients with advanced HCV-related cirrhosis. METHODS We carried out a multicenter longitudinal study in 31 patients with advanced hepatitis C virus (HCV)-related cirrhosis. We performed a non-targeted metabolomic analysis using gas chromatography-mass spectrometry and liquid chromatography-mass spectrometry, as well as analysis of inflammation-related biomarkers using Luminex technology. The statistical analysis was performed by Generalised Linear Mixed-effects Models (GLMM), correcting for multiple testing. RESULTS We found that increases of 2,3-butanediol (AMR = 1.15; q-value = 0.023) and taurocholic acid (AMR = 1.06; q-value < 0.001) were significantly associated with increases in HVPG and inflammatory biomarker levels from before DAA therapy to one year after completion of successful HCV treatment. CONCLUSIONS These metabolites have a potential role as indicators of portal hypertension evolution.
Collapse
Affiliation(s)
- Ana Virseda-Berdices
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rubén Martín-Escolano
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juan Berenguer
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Unidad de Enfermedades Infecciosas/VIH; Hospital General Universitario "Gregorio Marañón", Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Juan González-García
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Servicio de Medicina Interna-Unidad de VIH. Hospital Universitario La Paz, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | - Oscar Brochado-Kith
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - David Rojo
- Centro de Metabolómica y Bioanálisis (CEMBIO), Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, Urbanización Montepríncipe, Madrid, Spain
| | - Cristina Díez
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Servicio de Medicina Interna-Unidad de VIH. Hospital Universitario La Paz, Madrid, Spain
| | - Víctor Hontañon
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Servicio de Medicina Interna-Unidad de VIH. Hospital Universitario La Paz, Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | - Leire Pérez-Latorre
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Unidad de Enfermedades Infecciosas/VIH; Hospital General Universitario "Gregorio Marañón", Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Luis Ibañez-Samaniego
- Servicio de Aparato Digestivo, Hospital General Universitario 'Gregorio Marañón', Madrid, Spain
| | - Elba Llop-Herrera
- Departamento de Gastroenterología, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Antonio Olveira
- Servicio de Aparato Digestivo, Hospital Universitario La Paz, Madrid, Spain
| | - Amanda Fernández-Rodríguez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, Urbanización Montepríncipe, Madrid, Spain
| | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Ángeles Jiménez-Sousa
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
3
|
Campos-Murguia A, Hupa-Breier KL, Hartleben B, Wedemeyer H, Taubert R, Engel B. Elevated Plasma CXCL8 Concentrations in Significant Fibrosis but Not in Subclinical Rejection After Adult Liver Transplantation. Transplant Direct 2024; 10:e1592. [PMID: 39877649 PMCID: PMC11774564 DOI: 10.1097/txd.0000000000001592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/31/2025] Open
Abstract
Background The noninvasive detection of subclinical graft injury including subclinical T cell-mediated rejection (subTCMR) is one of the unresolved challenges after liver transplantation. Recently, serum C-X-C motif chemokine ligand 8 (CXCL8) was proposed as a highly accurate marker of subTCMR in pediatric liver transplant recipients. We aimed to evaluate the accuracy of the quantification of this chemokine for predicting subTCMR in adult liver transplant recipients, as well as its capacity to classify patients who could benefit from immunosuppression reduction. Methods Plasma CXCL8 concentrations were measured retrospectively in a prospectively collected cohort of adult liver transplant recipients with well-characterized histologic phenotypes. Results In total, 78 patients were included. Median plasma CXCL8 concentrations did not differ (P = 0.24) between patients without histological evidence of rejection (3.6 [0.4-22.0] pg/mL), subTCMR (11.5 [0.4-41.0] pg/mL), clinical TCMR (9.4 [0.4-40.5] pg/mL), and other etiologies of graft injury (8.7 [0.4-31.2] pg/mL). Likewise, plasma CXCL8 concentrations did not discriminate between patients within and outside histologic criteria for immunosuppression reduction that were proposed by the 2016 Banff Working Group on Liver Allograft Pathology (cutoff: 10.9 pg/mL, sensitivity: 0.48, and specificity: 0.79). Furthermore, weak correlation was found between plasma CXCL8 and alanine aminotransferase and aspartate aminotransferase (Spearman ρ = 0.18 and 0.25). Patients with significant fibrosis (17.8 [0.4-40.5] pg/mL) showed higher plasma CXCL8 concentrations than patients without fibrosis (8.2 [0.4-41.0] pg/mL; P = 0.05). Conclusions Plasma CXCL8 concentrations are not predictive of subclinical graft injury or of histological criteria for the minimization of immunosuppression in adult liver transplant recipients.
Collapse
Affiliation(s)
- Alejandro Campos-Murguia
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Katharina Luise Hupa-Breier
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Björn Hartleben
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Bastian Engel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
Raabe J, Kaiser KM, ToVinh M, Finnemann C, Lutz P, Hoffmeister C, Bischoff J, Goeser F, Kaczmarek DJ, Glowka TR, Manekeller S, Charpentier A, Langhans B, Nischalke HD, Toma M, Strassburg CP, Spengler U, Abdallah AT, Krämer B, Nattermann J. Identification and characterization of a hepatic IL-13-producing ILC3-like population potentially involved in liver fibrosis. Hepatology 2023; 78:787-802. [PMID: 37029085 DOI: 10.1097/hep.0000000000000350] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/12/2022] [Indexed: 04/09/2023]
Abstract
BACKGROUND AND AIMS Human innate lymphoid cells (ILCs) are critically involved in the modulation of homeostatic and inflammatory processes in various tissues. However, only little is known about the composition of the intrahepatic ILC pool and its potential role in chronic liver disease. Here, we performed a detailed characterization of intrahepatic ILCs in both healthy and fibrotic livers. APPROACH AND RESULTS A total of 50 livers (nonfibrotic = 22, and fibrotic = 29) were analyzed and compared with colon and tonsil tissue (each N = 14) and peripheral blood (N = 32). Human intrahepatic ILCs were characterized ex vivo and on stimulation using flow cytometry and single-cell RNA sequencing. ILC differentiation and plasticity were analyzed by both bulk and clonal expansion experiments. Finally, the effects of ILC-derived cytokines on primary human HSteCs were studied. Unexpectedly, we found that an "unconventional" ILC3-like cell represented the major IL-13-producing liver ILC subset. IL-13 + ILC3-like cells were specifically enriched in the human liver, and increased frequencies of this cell type were found in fibrotic livers. ILC3-derived IL-13 production induced upregulation of proinflammatory genes in HSteCs, indicating a potential role in the regulation of hepatic fibrogenesis. Finally, we identified KLRG1-expressing ILC precursors as the potential progenitor of hepatic IL-13 + ILC3-like cells. CONCLUSIONS We identified a formerly undescribed subset of IL-13-producing ILC3-like cells that is enriched in the human liver and may be involved in the modulation of chronic liver disease.
Collapse
Affiliation(s)
- Jan Raabe
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Kim M Kaiser
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Michael ToVinh
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Claudia Finnemann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Philipp Lutz
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | | | - Jenny Bischoff
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Felix Goeser
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | | | - Tim R Glowka
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Arthur Charpentier
- Department of Otorhinolaryngology, University Hospital Bonn, Bonn, Germany
| | - Bettina Langhans
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | | | - Marieta Toma
- Department of Pathology, University Hospital Bonn, Bonn, Germany
| | | | - Ulrich Spengler
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Ali T Abdallah
- Interdisciplinary Center for Clinical Research, RWTH Aachen University, Aachen, Germany
- The German Center for Infection Research (DZIF)
| | - Benjamin Krämer
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
- The German Center for Infection Research (DZIF)
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
- The German Center for Infection Research (DZIF)
| |
Collapse
|
5
|
Godbole N, Nyholm I, Hukkinen M, Davidson JR, Tyraskis A, Eloranta K, Andersson N, Lohi J, Heikkilä P, Kyrönlahti A, Pihlajoki M, Davenport M, Heikinheimo M, Pakarinen MP. Prognostic and Pathophysiologic Significance of IL-8 (CXCL8) in Biliary Atresia. J Clin Med 2021; 10:jcm10122705. [PMID: 34207442 PMCID: PMC8234515 DOI: 10.3390/jcm10122705] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-8 (CXCL8), a chemokine involved in neutrophil recruitment, has been implicated in ductular reaction and liver fibrogenesis. We studied liver and serum IL-8 expression in a large biliary atresia (BA) cohort and explored its prognostic and pathophysiological potential. IL-8 expression was assessed in liver utilizing quantitative polymerase chain reaction (qPCR), immunohistochemistry and in situ hybridization and in serum using an enzyme-linked immunosorbent assay, among 115 BA patients, 10 disease controls and 68 normal controls. Results were correlated to portoenterostomy (PE) outcomes, biochemical and histological liver injury, transcriptional markers of fibrosis and cholangiocytes, and expression of other related cytokines. IL-8 was markedly overexpressed in liver and serum of BA patients at PE (n = 88) and in serum samples obtained during postoperative follow-up (n = 40). IL-8 expression in the liver was predominantly in cholangiocytes within areas of ductular reaction. Liver IL-8 mRNA expression correlated positively with its serum concentration, bile ductular proliferation, Metavir fibrosis stage, and transcriptional markers of activated myofibroblasts (ACTA2) and cholangiocytes (KRT19). Taken together, IL-8 may mediate liver injury in BA by promoting ductular reaction and associated liver fibrogenesis. Prognostic value of serum IL-8 to predict native liver survival was limited and confined to the postoperative period after PE.
Collapse
Affiliation(s)
- Nimish Godbole
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group and Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland
| | - Iiris Nyholm
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group and Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland
| | - Maria Hukkinen
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group and Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland
| | - Joseph R. Davidson
- Department of Pediatric Surgery, GOS-UCL Institute of Child Health, London WC1N 1EH, UK;
- Department of Pediatric Surgery, King’s College Hospital, London SE5 9RS, UK; (A.T.); (M.D.)
| | - Athanasios Tyraskis
- Department of Pediatric Surgery, King’s College Hospital, London SE5 9RS, UK; (A.T.); (M.D.)
| | - Katja Eloranta
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
| | - Noora Andersson
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
| | - Jouko Lohi
- Department of Pathology, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (J.L.); (P.H.)
| | - Päivi Heikkilä
- Department of Pathology, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (J.L.); (P.H.)
| | - Antti Kyrönlahti
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
| | - Marjut Pihlajoki
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
| | - Mark Davenport
- Department of Pediatric Surgery, King’s College Hospital, London SE5 9RS, UK; (A.T.); (M.D.)
| | - Markku Heikinheimo
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Mikko P. Pakarinen
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland; (N.G.); (I.N.); (M.H.); (K.E.); (N.A.); (A.K.); (M.P.); (M.H.)
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group and Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland
- Correspondence:
| |
Collapse
|
6
|
Abouelasrar Salama S, Gouwy M, De Zutter A, Pörtner N, Vanbrabant L, Berghmans N, De Buck M, Struyf S, Van Damme J. Induction of Chemokines by Hepatitis C Virus Proteins: Synergy of the Core Protein with Interleukin-1β and Interferon-γ in Liver Bystander Cells. J Interferon Cytokine Res 2020; 40:195-206. [PMID: 32031878 DOI: 10.1089/jir.2019.0115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection accounts for a large proportion of hepatic fibrosis and carcinoma cases observed worldwide. Mechanisms involved in HCV-induced hepatic injury have yet to be fully elucidated. Of particular interest is the capacity of HCV to regulate inflammatory responses. Here, we reveal modulation of cytokine activity by the HCV proteins non-structural protein 3 (NS3), glycoprotein E2, and core protein for their ability to induce chemokine expression in various liver bystander cells. Chemokines sustain chronic liver inflammation and relay multiple fibrogenic effects. CCL2, CCL3, CCL20, CXCL8, and CXCL10 were differentially expressed after treatment of monocytes, fibroblasts, or liver sinusoidal microvascular endothelial cells (LSECs) with HCV proteins. In comparison to NS3 and glycoprotein E2, core protein was a stronger inducer of chemokines in liver bystander cells. Interferon-γ (IFN-γ) and interleukin-1β (IL-1β) synergized with core protein to induce CCL2, CCL20, CXCL8, or CXCL10 in fibroblasts or LSECs. These findings reveal new mechanisms of hepatic injury caused by HCV.
Collapse
Affiliation(s)
- Sara Abouelasrar Salama
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Alexandra De Zutter
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Noëmie Pörtner
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Lotte Vanbrabant
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Nele Berghmans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Mieke De Buck
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Meier RPH, Meyer J, Montanari E, Lacotte S, Balaphas A, Muller YD, Clément S, Negro F, Toso C, Morel P, Buhler LH. Interleukin-1 Receptor Antagonist Modulates Liver Inflammation and Fibrosis in Mice in a Model-Dependent Manner. Int J Mol Sci 2019; 20:1295. [PMID: 30875826 PMCID: PMC6471711 DOI: 10.3390/ijms20061295] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Interleukin-1 (IL-1)β and IL-1 receptor antagonist (IL-1Ra) have been proposed as important mediators during chronic liver diseases. We aimed to determine whether the modulation of IL-1β signaling with IL-1Ra impacts on liver fibrosis. METHODS We assessed the effects of IL-1β on human hepatic stellate cells (HSC) and in mouse models of liver fibrosis induced by bile duct ligation (BDL) or carbon tetrachloride treatment (CCl-4). RESULTS Human HSCs treated with IL-1β had increased IL-1β, IL-1Ra, and MMP-9 expressions in vitro. HSCs treated with IL-1β had reduced α-smooth muscle actin expression. These effects were all prevented by IL-1Ra treatment. In the BDL model, liver fibrosis and Kuppfer cell numbers were increased in IL-1Ra KO mice compared to wild type mice and wild type mice treated with IL-1Ra. In contrast, after CCl-4 treatment, fibrosis, HSC and Kupffer cell numbers were decreased in IL-1Ra KO mice compared to the other groups. IL-1Ra treatment provided a modest protective effect in the BDL model and was pro-fibrotic in the CCl-4 model. CONCLUSIONS We demonstrated bivalent effects of IL-1Ra during liver fibrosis in mice. IL-1Ra was detrimental in the CCl-4 model, whereas it was protective in the BDL model. Altogether these data suggest that blocking IL-1-mediated inflammation may be beneficial only in selective liver fibrotic disease.
Collapse
Affiliation(s)
- Raphael P H Meier
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
- Transplant Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Jeremy Meyer
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Elisa Montanari
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Stephanie Lacotte
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Alexandre Balaphas
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Yannick D Muller
- Transplant Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Sophie Clément
- Division of Clinical Pathology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Francesco Negro
- Division of Clinical Pathology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
- Division of Gastroenterology and Hepatology, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Christian Toso
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Philippe Morel
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| | - Leo H Buhler
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospitals and Medical School, 1211 Geneva, Switzerland.
| |
Collapse
|
8
|
Franko A, Kovarova M, Feil S, Feil R, Wagner R, Heni M, Königsrainer A, Ruoß M, Nüssler AK, Weigert C, Häring HU, Lutz SZ, Peter A. cGMP-dependent protein kinase I (cGKI) modulates human hepatic stellate cell activation. Metabolism 2018; 88:22-30. [PMID: 30195474 DOI: 10.1016/j.metabol.2018.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/23/2018] [Accepted: 09/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The activation of hepatic stellate cells (HSCs) plays a crucial role in liver fibrosis, however the role of HSCs is less understood in hepatic insulin resistance. Since in the liver cGMP-dependent protein kinase I (cGKI) was detected in HSC but not in hepatocytes, and cGKI-deficient mice that express cGKI selectively in smooth muscle but not in other cell types (cGKI-SM mice) displayed hepatic insulin resistance, we hypothesized that cGKI modulates HSC activation and insulin sensitivity. MATERIALS AND METHODS To study stellate cell activation in cGKI-SM mice, retinol storage and gene expression were studied. Moreover, in the human stellate cell line LX2, the consequences of cGKI-silencing on gene expression were investigated. Finally, cGKI expression was examined in human liver biopsies covering a wide range of liver fat content. RESULTS Retinyl-ester concentrations in the liver of cGKI-SM mice were lower compared to wild-type animals, which was associated with disturbed expression of genes involved in retinol metabolism and inflammation. cGKI-silenced LX2 cells showed an mRNA expression profile of stellate cell activation, altered matrix degradation and activated chemokine expression. On the other hand, activation of LX2 cells suppressed cGKI expression. In accordance with this finding, in human liver biopsies, we observed a negative correlation between cGKI mRNA and liver fat content. CONCLUSIONS These results suggest that the lack of cGKI possibly leads to stellate cell activation, which stimulates chemokine expression and activates inflammatory processes, which could disturb hepatic insulin sensitivity.
Collapse
Affiliation(s)
- Andras Franko
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Marketa Kovarova
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany
| | - Robert Wagner
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Martin Heni
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
| | - Marc Ruoß
- Department of Traumatology, BG Trauma Clinic, Siegfried Weller Institute for Trauma Research, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Clinic, Siegfried Weller Institute for Trauma Research, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Cora Weigert
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Stefan Z Lutz
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Andreas Peter
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University Hospital Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str 10, 72076 Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
9
|
Karimi-Sales E, Jeddi S, Ebrahimi-Kalan A, Alipour MR. trans-Chalcone prevents insulin resistance and hepatic inflammation and also promotes hepatic cholesterol efflux in high-fat diet-fed rats: modulation of miR-34a-, miR-451-, and miR-33a-related pathways. Food Funct 2018; 9:4292-4298. [PMID: 30039136 DOI: 10.1039/c8fo00923f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Insulin resistance and inflammation are strongly linked to non-alcoholic fatty liver disease (NAFLD) as a feature of the metabolic syndrome. Furthermore, the role of dysregulation of miR-34a, miR-451, and miR-33a in pathogenesis and progression of NAFLD has been identified. trans-Chalcone is a simple chalcone with anti-diabetic and anti-inflammatory activities. However, to the best of our knowledge, miRNA-dependent mechanisms of these protective effects under pathologic conditions are not understood. Thus, this study, for the first time, aimed to evaluate the effects of trans-Chalcone on miR-34a, miR-451, and miR-33a signaling pathways in the liver of high-fat (HF) emulsion-fed rats. To this aim, twenty-one rats were randomly and equally divided into three groups: control, which was gavaged with 10% tween 80; HF, which was gavaged with HF emulsion and 10% tween 80; and HF + trans-Chalcone (HF + TC), which was gavaged with HF emulsion and trans-Chalcone. Then, circulating levels of glucose and insulin were measured and used for the calculation of HOMA-IR. Hepatic expression levels of miR-34a, miR-451, miR-33a, SIRT1, and ABCA1 and also protein levels of ABCA1 and IL-8 were assayed. In this study, trans-chalcone increased hepatic cholesterol efflux and prevented insulin resistance and liver inflammation in HF emulsion-fed rats. These protective effects were modulated through the down-regulation of miR-34a and its associated elevation of SIRT1, the up-regulation of miR-451 which was associated with a reduction in IL-8, and the inhibition of miR-33a which was related to the elevation of ABCA1 in the liver of HF emulsion-fed rats. Therefore, trans-Chalcone exerts its beneficial effects by targeting hepatic miR-34a-, miR-451-, and miR-33a-related pathways.
Collapse
Affiliation(s)
- Elham Karimi-Sales
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | | | | |
Collapse
|
10
|
Morozov VA, Lagaye S. Hepatitis C virus: Morphogenesis, infection and therapy. World J Hepatol 2018; 10:186-212. [PMID: 29527256 PMCID: PMC5838439 DOI: 10.4254/wjh.v10.i2.186] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/11/2018] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is a major cause of liver diseases including liver cirrhosis and hepatocellular carcinoma. Approximately 3% of the world population is infected with HCV. Thus, HCV infection is considered a public healthy challenge. It is worth mentioning, that the HCV prevalence is dependent on the countries with infection rates around 20% in high endemic countries. The review summarizes recent data on HCV molecular biology, the physiopathology of infection (immune-mediated liver damage, liver fibrosis and lipid metabolism), virus diagnostic and treatment. In addition, currently available in vitro, ex vivo and animal models to study the virus life cycle, virus pathogenesis and therapy are described. Understanding of both host and viral factors may in the future lead to creation of new approaches in generation of an efficient therapeutic vaccine.
Collapse
Affiliation(s)
- Vladimir Alexei Morozov
- Center for HIV and Retrovirology, Department of Infectious Diseases, Robert Koch Institute, Berlin 13353, Germany
| | - Sylvie Lagaye
- Department of Immunology, Institut Pasteur, INSERM U1223, Paris 75015, France
| |
Collapse
|
11
|
Cheng JC, Tseng CP, Liao MH, Peng CY, Yu JS, Chuang PH, Huang JT, Chen JJW. Activation of hepatic stellate cells by the ubiquitin C-terminal hydrolase 1 protein secreted from hepatitis C virus-infected hepatocytes. Sci Rep 2017; 7:4448. [PMID: 28667290 PMCID: PMC5493679 DOI: 10.1038/s41598-017-04259-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/11/2017] [Indexed: 12/17/2022] Open
Abstract
Hepatitis C virus (HCV) infection of hepatocytes promotes liver fibrosis by activation of hepatic stellate cells (HSCs) and excessive deposition of extracellular matrix in liver tissue. Whether or not host factors released from the HCV-infected hepatocytes play role in HSCs activation is unclear. In this study, HSCs were activated by the conditioned medium derived from HCV replicon cells. Secretomic profiling of HCV replicon cells and the parental Huh7 cells revealed ubiquitin carboxy-terminal hydrolase L1 (UCHL1) as a novel secreted protein from HCV-infected hepatocytes. UCHL1 expression in hepatocytes was induced by HCV infection. UCHL1 was expressed in the liver and found in the plasma of patients with chronic hepatitis C. Molecular analysis by use of the anti-UCHL1 neutralization antibody and purified UCHL1 protein showed that secreted UCHL1 protein was bound to the cell surface of HSCs and activated JNK signaling leading to overexpression of alpha-smooth muscle actin and the activation of HSCs. These results provide further for understanding the underlying mechanism in HCV-mediated hepatic fibrogenesis.
Collapse
Affiliation(s)
- Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 40402, Taiwan.
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan.,Molecular Medicine Research Center, Chang Gung University, Taoyuan, 33302, Taiwan.,Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, 33302, Taiwan
| | - Mei-Huei Liao
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 40402, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, 40402, Taiwan
| | - Cheng-Yuan Peng
- Department of Internal Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Jau-Song Yu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan.,Molecular Medicine Research Center, Chang Gung University, Taoyuan, 33302, Taiwan.,Liver Research Center, Chang Gung Memorial Hospital, Linkou, 33302, Taiwan
| | - Po-Heng Chuang
- Department of Internal Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Jing-Tang Huang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 40402, Taiwan
| | - Jeremy J W Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, 40402, Taiwan
| |
Collapse
|
12
|
Horas H Nababan S, Nishiumi S, Kawano Y, Kobayashi T, Yoshida M, Azuma T. Adrenic acid as an inflammation enhancer in non-alcoholic fatty liver disease. Arch Biochem Biophys 2017; 623-624:64-75. [DOI: 10.1016/j.abb.2017.04.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/23/2017] [Accepted: 04/25/2017] [Indexed: 12/19/2022]
|
13
|
Tan J, Wu J. Current progress in understanding the molecular pathogenesis of burn scar contracture. BURNS & TRAUMA 2017; 5:14. [PMID: 28546987 PMCID: PMC5441009 DOI: 10.1186/s41038-017-0080-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/17/2017] [Indexed: 01/17/2023]
Abstract
Abnormal wound healing is likely to induce scar formation, leading to dysfunction, deformity, and psychological trauma in burn patients. Despite the advancement of medical care treatment, scar contracture in burn patients remains a challenge. Myofibroblasts play a key role in scar contracture. It has been demonstrated that myofibroblasts, as well as inflammatory cells, fibroblasts, endothelial cells, and epithelial cells, secrete transforming growth factor-β1 (TGF-β1) and other cytokines, which can promote persistent myofibroblast activation via a positive regulation loop. In addition to the cellular contribution, the microenvironments, including the mechanical tension and integrin family, are also involved in scar contracture. Most recently, eukaryotic initiation factor 6 (eIF6), an upstream regulator of TGF-β1, has been demonstrated to be involved in myofibroblast differentiation and contraction in both in vitro fibroblast-populated collagen lattice (FPCL) and in vivo external mechanical stretch models. Moreover, the data showed that P311 could induce the transdifferentiation of epidermal stem cells to myofibroblasts by upregulating TGF-β1 expression, which mediated myofibroblast contraction. In this review, we briefly described the most current progress on the biological function of myofibroblasts in scar contracture and subsequently summarized the molecular events that initiated contracture. This would help us better understand the molecular basis of scar contracture as well as to find a comprehensive strategy for preventing/managing scar contracture.
Collapse
Affiliation(s)
- Jianglin Tan
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injuries, Chongqing Key Laboratory for Disease Proteomics, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Jun Wu
- Department of Burns, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| |
Collapse
|
14
|
Ajmera V, Perito ER, Bass NM, Terrault NA, Yates KP, Gill R, Loomba R, Diehl AM, Aouizerat B. Novel plasma biomarkers associated with liver disease severity in adults with nonalcoholic fatty liver disease. Hepatology 2017; 65:65-77. [PMID: 27532276 PMCID: PMC5191932 DOI: 10.1002/hep.28776] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 06/02/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED Despite the high prevalence of nonalcoholic fatty liver disease (NAFLD), therapeutic options and noninvasive markers of disease activity and severity remain limited. We investigated the association between plasma biomarkers and liver histology in order to identify markers of disease activity and severity in patients with biopsy-proven NAFLD. Thirty-two plasma biomarkers chosen a priori as possible discriminators of NAFLD were measured in participants enrolled in the Nonalcoholic Steatohepatitis (NASH) Clinical Research Network. Dichotomized histologic outcomes were evaluated using centrally read biopsies. Biomarkers with statistically significant associations with NAFLD histology were evaluated in multivariable models adjusted for clinical factors. Of 648 participants (74.4% white, 61.7% female, mean age 47.7 years), 58.0% had definite NASH, 55.5% had mild/no fibrosis (stage 0-1), and 44.4% had significant fibrosis (stage 2-4). Increased activated plasminogen activator inhibitor 1 had a strong association with definite NASH compared to not NASH or borderline NASH in multivariable analysis (odds ratio = 1.20, 95% confidence interval 1.08-1.34, P < 0.001). Biomarkers associated with significant fibrosis (versus mild/no fibrosis) in multivariable analysis included higher levels of interleukin-8, monocyte chemoattractant protein-1, resistin, soluble interleukin-1 receptor I, soluble interleukin-2 receptor alpha, and tumor necrosis factor alpha and lower levels of insulin-like growth factor 2. CONCLUSIONS Specific plasma biomarkers are significantly associated with disease activity and severity of fibrosis in NAFLD and are potentially valuable tools for noninvasive stratification of patients with NAFLD and identification of targets for therapeutic intervention. (Hepatology 2017;65:65-77).
Collapse
Affiliation(s)
- Veeral Ajmera
- Gastroenterology, UCSF, San Francisco, CA, United States
| | - Emily R. Perito
- Pediatric Gastroenterology, UCSF, San Francisco, CA, United States
| | - Nathan M. Bass
- Gastroenterology, UCSF, San Francisco, CA, United States
| | | | | | - Ryan Gill
- Pathology, UCSF, San Francisco, CA, United States
| | - Rohit Loomba
- Gastroenterology, UCSD, San Diego, CA, United States
| | - Anna Mae Diehl
- Gastroenterology, Duke University, Durham, NC, United States
| | | | | |
Collapse
|
15
|
Hepatic SATB1 induces paracrine activation of hepatic stellate cells and is upregulated by HBx. Sci Rep 2016; 6:37717. [PMID: 27883059 PMCID: PMC5121621 DOI: 10.1038/srep37717] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/28/2016] [Indexed: 12/23/2022] Open
Abstract
Chronic hepatitis B virus (HBV) infection is a major cause of chronic liver diseases, but its involvement in hepatic fibrogenesis remains unclear. Special AT-rich binding protein 1 (SATB1) has been implicated in reprogramming chromatin organization and transcription profiles in many cancers and non-cancer-related conditions. We found that hepatic SATB1 expression was significantly up-regulated in fibrotic tissues from chronic hepatitis B virus (HBV)-infected patients and HBV transgenic (HBV-Tg) mouse model. Knockdown of SATB1 in the liver significantly alleviated CCl4-induced fibrosis in HBV-Tg mouse model. Moreover, we suggested HBV encoded x protein (HBx) induced SATB1 expression through activation of JNK and ERK pathways. Enforced expression of SATB1 in hepatocytes promoted the activation and proliferation of hepatic stellate cells (HSCs) by secretion of connective tissue growth factor (CTGF), Interleukin-6 (IL-6) and platelet derived growth factor-A (PDGF-AA). Our findings demonstrated that HBx upregulated hepatic SATB1 which exerted pro-fibrotic effects by paracrine activation of stellate cells in HBV-related fibrosis.
Collapse
|
16
|
Zweers SJ, Shiryaev A, Komuta M, Vesterhus M, Hov JR, Perugorria MJ, de Waart DR, Chang JC, Tol S, Te Velde AA, de Jonge WJ, Banales JM, Roskams T, Beuers U, Karlsen TH, Jansen PL, Schaap FG. Elevated interleukin-8 in bile of patients with primary sclerosing cholangitis. Liver Int 2016; 36:1370-7. [PMID: 26866350 DOI: 10.1111/liv.13092] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/30/2016] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS To better understand the pathogenesis of primary sclerosing cholangitis, anti- and pro-inflammatory factors were studied in bile. METHODS Ductal bile of PSC patients (n = 36) and controls (n = 20) was collected by endoscopic retrograde cholangiography. Gallbladder bile was collected at liver transplantation. Bile samples were analysed for cytokines, FGF19 and biliary lipids. Hepatobiliary tissues of PSC and non-PSC patients (n = 8-11 per patient group) were collected at transplantation and were analysed for IL8 and FGF19 mRNA expression and IL8 localization. The effect of IL8 on proliferation of primary human cholangiocytes and expression of pro-fibrotic genes was studied. RESULTS In PSC patients, median IL8 in ductal bile was 6.6 ng/ml vs. 0.24 ng/ml in controls. Median IL8 in gallbladder bile was 7.6 ng/ml in PSC vs. 2.2 and 0.3 ng/ml in two control groups. IL8 mRNA in PSC gallbladder was increased and bile ducts stained positive for IL8. In vitro, IL8 induced proliferation of primary human cholangiocytes and increased the expression of pro-fibrotic genes. CONCLUSION Elevation of IL8 in bile of PSC patients, collected at different stages of disease, indicates an ongoing inflammatory stimulus that drives IL8 production. This challenges the idea that advanced PSC is a burned-out disease, and calls for reconsideration of anti-inflammatory therapy in PSC.
Collapse
Affiliation(s)
- Serge J Zweers
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Alexey Shiryaev
- Division of Cancer Medicine, Surgery and Transplantation, Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Cancer Medicine, Surgery and Transplantation, Research Institute of Internal Medicine, K.G. Jebsen Inflammation Research Center, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Mina Komuta
- Morphology and Molecular Pathology, University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Mette Vesterhus
- Division of Cancer Medicine, Surgery and Transplantation, Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway.,National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Johannes R Hov
- Division of Cancer Medicine, Surgery and Transplantation, Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Cancer Medicine, Surgery and Transplantation, Research Institute of Internal Medicine, K.G. Jebsen Inflammation Research Center, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - María J Perugorria
- Department of Liver and Department of Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastián, Spain
| | - D Rudi de Waart
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Jung-Chin Chang
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Shanna Tol
- Department of Surgery, Academic Medical Center, Amsterdam, The Netherlands
| | - Anje A Te Velde
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Jesus M Banales
- Department of Liver and Department of Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastián, Spain
| | - Tania Roskams
- Morphology and Molecular Pathology, University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Ulrich Beuers
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Tom H Karlsen
- Division of Cancer Medicine, Surgery and Transplantation, Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Cancer Medicine, Surgery and Transplantation, Research Institute of Internal Medicine, K.G. Jebsen Inflammation Research Center, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Peter L Jansen
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands.,Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.,Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Frank G Schaap
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands.,Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
17
|
Gu YJ, Sun WY, Zhang S, Li XR, Wei W. Targeted blockade of JAK/STAT3 signaling inhibits proliferation, migration and collagen production as well as inducing the apoptosis of hepatic stellate cells. Int J Mol Med 2016; 38:903-11. [PMID: 27460897 DOI: 10.3892/ijmm.2016.2692] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 07/08/2016] [Indexed: 11/06/2022] Open
Abstract
Protein tyrosine kinases belonging to the Janus kinase (JAK) family are associated with many cytokine receptors, which, on ligand binding, regulate important cellular functions such as proliferation, apoptosis and differentiation. The protective effects of JAK inhibitors on fibrotic diseases such as myelofibrosis and bone marrow fibrosis have been demonstrated in previous studies. The JAK inhibitor SHR0302 is a synthetic molecule that potently inhibits all members of the JAK family, particularly JAK1. However, its effect on hepatic fibrosis has not been investigated to date, to the best of our knowledge. In the present study, the effects of SHR0302 on the activation, proliferation, migration and apoptosis of hepatic stellate cells (HSCs) as well as HSC collagen production were investigated. Our data demonstrated that treatment with SHR0302 (10-9-10-5 mol/l) exerted an inhibitory effect on the activation, proliferation and migration of HSCs. In addition, the expression of collagen I and collagen III were significantly decreased following treatment with SHR0302. Furthermore, SHR0302 induced the apoptosis of HSCs, which was demonstrated by Annexin V/PI staining. SHR0302 significantly increased the activation of caspase-3 and Bax in HSCs whereas it decreased the expression of Bcl-2. SHR0302 also inhibited the activation of Akt signaling pathway. The pharmacological inhibition of the JAK1/signal transducer and activator of transcription (STAT)3 pathway led to the disruption of functions essential for HSC growth. Taken together, these findings provide evidence that SHR0302 may have the potential to alleviate hepatic fibrosis by targeting HSC functions.
Collapse
Affiliation(s)
- Yuan-Jing Gu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Sen Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Xin-Ran Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
18
|
Mattoli S. Involvement of fibrocytes in asthma and clinical implications. Clin Exp Allergy 2016; 45:1497-509. [PMID: 25752439 DOI: 10.1111/cea.12525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bloodborne fibrocytes are bone marrow-derived cells that participate in immune responses and exhibit pro-inflammatory and matrix remodelling properties. In patients with asthma receiving an adequate treatment, the blood fibrocyte count is very low and comparable to that obtained in healthy individuals. In these patients, a transient increase in fibrocyte numbers in the peripheral blood and in the airways occurs in concomitance with increased bronchial inflammation and reflects disease worsening and the need for more intensive treatment. Persistently elevated numbers of fibrocytes in the peripheral blood and in the bronchial mucosa are observed in chronically undertreated or corticosteroid-resistant asthma and are associated with persistent airway inflammation and ongoing remodelling of the bronchial wall. The asthmatic bronchial epithelium is the main source of fibrocyte chemoattractants in asthma and contributes with T helper type 2 lymphocytes and eosinophils to promote the proliferation and pro-remodelling function of recruited fibrocytes. The presence of elevated numbers of fibrocytes in the bronchial mucosa of allergic patients with undertreated or treatment-resistant asthma may also increase the risk of acute exacerbations because these cells can amplify T helper type 2 lymphocyte-driven inflammation on every exposure to the clinically relevant allergen and can promote further inflammation on rhinovirus infections by allowing viral replication and releasing additional pro-inflammatory factors. Improved methods for the isolation and functional analysis of pure populations of viable circulating fibrocytes have allowed a better understanding of the effector role of these cells. A reliable and clinically applicable assay has been developed to measure blood fibrocyte counts as outcome measure in future clinical trials. New therapeutic agents are needed to block both persistent inflammation and fibrocytosis in corticosteroid-resistant asthma.
Collapse
Affiliation(s)
- S Mattoli
- Avail Biomedical Research Institute, Scientific Direction and Project Management Centre, Basel, Switzerland
| |
Collapse
|
19
|
Zhang T, Tseng C, Zhang Y, Sirin O, Corn PG, Li-Ning-Tapia EM, Troncoso P, Davis J, Pettaway C, Ward J, Frazier ML, Logothetis C, Kolonin MG. CXCL1 mediates obesity-associated adipose stromal cell trafficking and function in the tumour microenvironment. Nat Commun 2016; 7:11674. [PMID: 27241286 PMCID: PMC4895055 DOI: 10.1038/ncomms11674] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/19/2016] [Indexed: 12/25/2022] Open
Abstract
White adipose tissue (WAT) overgrowth in obesity is linked with increased aggressiveness of certain cancers. Adipose stromal cells (ASCs) can become mobilized from WAT, recruited by tumours and promote cancer progression. Mechanisms underlying ASC trafficking are unclear. Here we demonstrate that chemokines CXCL1 and CXCL8 chemoattract ASC by signalling through their receptors, CXCR1 and CXCR2, in cell culture models. We further show that obese patients with prostate cancer have increased epithelial CXCL1 expression. Concomitantly, we observe that cells with ASC phenotype are mobilized and infiltrate tumours in obese patients. Using mouse models, we show that the CXCL1 chemokine gradient is required for the obesity-dependent tumour ASC recruitment, vascularization and tumour growth promotion. We demonstrate that αSMA expression in ASCs is induced by chemokine signalling and mediates the stimulatory effects of ASCs on endothelial cells. Our data suggest that ASC recruitment to tumours, driven by CXCL1 and CXCL8, promotes prostate cancer progression. Adipose stromal cells (ASC) have been shown to migrate to tumours and promote tumour growth. Using animal models and human tissue samples, the authors show here that ASC recruitment to prostate cancers is mediated by the chemokine CXCL1, which is secreted from tumour cells, and acts on CXCR1 on ASCs.
Collapse
Affiliation(s)
- Tao Zhang
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Chieh Tseng
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Yan Zhang
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Olga Sirin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Paul G Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Elsa M Li-Ning-Tapia
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John Davis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Curtis Pettaway
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John Ward
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Marsha L Frazier
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
20
|
Mustafa HN. The role of curcumin in streptozotocin-induced hepatic damage and the trans-differentiation of hepatic stellate cells. Tissue Cell 2016; 48:81-8. [DOI: 10.1016/j.tice.2016.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 01/19/2016] [Accepted: 02/07/2016] [Indexed: 10/22/2022]
|
21
|
Branche E, Conzelmann S, Parisot C, Bedert L, Lévy PL, Bartosch B, Clément S, Negro F. Hepatitis C Virus Increases Occludin Expression via the Upregulation of Adipose Differentiation-Related Protein. PLoS One 2016; 11:e0146000. [PMID: 26731658 PMCID: PMC4701191 DOI: 10.1371/journal.pone.0146000] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/11/2015] [Indexed: 12/12/2022] Open
Abstract
The hepatitis C virus (HCV) life cycle is closely associated with lipid metabolism. In particular, HCV assembly initiates at the surface of lipid droplets. To further understand the role of lipid droplets in HCV life cycle, we assessed the relationship between HCV and the adipose differentiation-related protein (ADRP), a lipid droplet-associated protein. Different steps of HCV life cycle were assessed in HCV-infected human Huh-7 hepatoma cells overexpressing ADRP upon transduction with a lentiviral vector. HCV infection increased ADRP mRNA and protein expression levels by 2- and 1.5-fold, respectively. The overexpression of ADRP led to an increase of (i) the surface of lipid droplets, (ii) the total cellular neutral lipid content (2.5- and 5-fold increase of triglycerides and cholesterol esters, respectively), (iii) the cellular free cholesterol level (5-fold) and (iv) the HCV particle production and infectivity (by 2- and 3.5-fold, respectively). The investigation of different steps of the HCV life cycle indicated that the ADRP overexpression, while not affecting the viral replication, promoted both virion egress and entry (~12-fold), the latter possibly via an increase of its receptor occludin. Moreover, HCV infection induces an increase of both ADRP and occludin expression. In HCV infected cells, the occludin upregulation was fully prevented by the ADRP silencing, suggesting a specific, ADRP-dependent mechanism. Finally, in HCV-infected human livers, occludin and ADRP mRNA expression levels correlated with each other. Alltogether, these findings show that HCV induces ADRP, which in turns appears to confer a favorable environment to viral spread.
Collapse
Affiliation(s)
- Emilie Branche
- Department of Immunology and Pathology, Faculty of Medicine, Geneva, Switzerland
| | - Stéphanie Conzelmann
- Department of Immunology and Pathology, Faculty of Medicine, Geneva, Switzerland
| | - Clotilde Parisot
- Department of Immunology and Pathology, Faculty of Medicine, Geneva, Switzerland
| | - Ludmila Bedert
- Department of Immunology and Pathology, Faculty of Medicine, Geneva, Switzerland
| | - Pierre L. Lévy
- Department of Immunology and Pathology, Faculty of Medicine, Geneva, Switzerland
| | - Birke Bartosch
- CRCL, INSERM U1052, CNRS 5286, University of Lyon, Lyon, France
| | - Sophie Clément
- Divisions of Clinical Pathology, University Hospital, Geneva, Switzerland
- * E-mail: (FN); (SC)
| | - Francesco Negro
- Divisions of Clinical Pathology, University Hospital, Geneva, Switzerland
- Gastroenterology and Hepatology, University Hospital, Geneva, Switzerland
- * E-mail: (FN); (SC)
| |
Collapse
|
22
|
Perez Aguilar RC, Honoré SM, Genta SB, Sánchez SS. Hepatic fibrogenesis and transforming growth factor/Smad signaling activation in rats chronically exposed to low doses of lead. J Appl Toxicol 2015; 34:1320-31. [PMID: 25493318 DOI: 10.1002/jat.2955] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lead is an important heavy metal pollutant in the environment. The nervous system, kidney and liver are the most susceptible organs to lead deposition, showing that this pollutant has no single target system. To examine the cellular and molecular mechanisms involved in their pathobiology of chronic lead at low-dose exposure in the liver, male Wistar rats were exposed to 0.06% lead acetate in drinking water every day for 4 months. At the end of the study, hepatic metal accumulation, morphology and function were examined. Immunochemical staining and Western blot analysis were performed to detect extracellular matrix proteins, α-smooth muscle actin and transforming growth factor (TGF)β1/Smad pathway expression. Results showed increased laminin, collagen IV and fibronectin, located at the perisinusoidal space. Phenotypic transformation of hepatic stellate cells into myofibroblast-like cells was evidenced at the ultrastructural level and a significant expression of α-smooth muscle actin in Disse’s space was observed. These findings were associated with a marked increase in TGFβ1/Smad2/3 signaling. Our data suggest that, chronically, exposure to low levels of lead could trigger the onset of a hepatic fibrogenic process through upregulated TGFβ1/Smad signaling.
Collapse
|
23
|
Dong R, Zheng S. Interleukin-8: A critical chemokine in biliary atresia. J Gastroenterol Hepatol 2015; 30:970-6. [PMID: 25611432 DOI: 10.1111/jgh.12900] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2015] [Indexed: 12/15/2022]
Abstract
Biliary atresia (BA) is characterized by periductular inflammation and fibrosis and is associated with the progressive obliteration of the bile ducts. The induction and maintenance of systemic and local inflammatory responses plays a pivotal role in this process. Interleukin-8 (IL-8) is an important mediator of inflammation and the immune response in human disease. IL-8 is overexpressed in BA, and its expression positively correlates with inflammation and liver fibrosis. In this review, we focus on the available evidence, recent insights, and future clinical and preclinical possibilities regarding the role of IL-8 in BA.
Collapse
Affiliation(s)
- Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | | |
Collapse
|
24
|
Weiskirchen R, Tacke F. Cellular and molecular functions of hepatic stellate cells in inflammatory responses and liver immunology. Hepatobiliary Surg Nutr 2015; 3:344-63. [PMID: 25568859 DOI: 10.3978/j.issn.2304-3881.2014.11.03] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/17/2014] [Indexed: 12/11/2022]
Abstract
The liver is a central immunological organ. Liver resident macrophages, Kupffer cells (KC), but also sinusoidal endothelial cells, dendritic cells (DC) and other immune cells are involved in balancing immunity and tolerance against pathogens, commensals or food antigens. Hepatic stellate cells (HSCs) have been primarily characterized as the main effector cells in liver fibrosis, due to their capacity to transdifferentiate into collagen-producing myofibroblasts (MFB). More recent studies elucidated the fundamental role of HSC in liver immunology. HSC are not only the major storage site for dietary vitamin A (Vit A) (retinol, retinoic acid), which is essential for proper function of the immune system. This pericyte further represents a versatile source of many soluble immunological active factors including cytokines [e.g., interleukin 17 (IL-17)] and chemokines [C-C motif chemokine (ligand) 2 (CCL2)], may act as an antigen presenting cell (APC), and has autophagy activity. Additionally, it responds to many immunological triggers via toll-like receptors (TLR) (e.g., TLR4, TLR9) and transduces signals through pathways and mediators traditionally found in immune cells, including the Hedgehog (Hh) pathway or inflammasome activation. Overall, HSC promote rather immune-suppressive responses in homeostasis, like induction of regulatory T cells (Treg), T cell apoptosis (via B7-H1, PDL-1) or inhibition of cytotoxic CD8 T cells. In conditions of liver injury, HSC are important sensors of altered tissue integrity and initiators of innate immune cell activation. Vice versa, several immune cell subtypes interact directly or via soluble mediators with HSC. Such interactions include the mutual activation of HSC (towards MFB) and macrophages or pro-apoptotic signals from natural killer (NK), natural killer T (NKT) and gamma-delta T cells (γδ T-cells) on activated HSC. Current directions of research investigate the immune-modulating functions of HSC in the environment of liver tumors, cellular heterogeneity or interactions promoting HSC deactivation during resolution of liver fibrosis. Understanding the role of HSC as central regulators of liver immunology may lead to novel therapeutic strategies for chronic liver diseases.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- 1 Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, 2 Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- 1 Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, 2 Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
25
|
Nishitsuji H, Funami K, Shimizu Y, Ujino S, Seya T, Shimotohno K. Hepatitis C Virus (HCV)-Induced Inflammation: The Role of Cross-Talk Between HCV-Infected Hepatocytes and Stellate Cells. INFLAMMATION AND IMMUNITY IN CANCER 2015:109-121. [DOI: 10.1007/978-4-431-55327-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
26
|
Loizides-Mangold U, Clément S, Alfonso-Garcia A, Branche E, Conzelmann S, Parisot C, Potma EO, Riezman H, Negro F. HCV 3a core protein increases lipid droplet cholesteryl ester content via a mechanism dependent on sphingolipid biosynthesis. PLoS One 2014; 9:e115309. [PMID: 25522003 PMCID: PMC4270764 DOI: 10.1371/journal.pone.0115309] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 11/21/2014] [Indexed: 12/22/2022] Open
Abstract
Hepatitis C virus (HCV) infected patients often develop steatosis and the HCV core protein alone can induce this phenomenon. To gain new insights into the pathways leading to steatosis, we performed lipidomic profiling of HCV core protein expressing-Huh-7 cells and also assessed the lipid profile of purified lipid droplets isolated from HCV 3a core expressing cells. Cholesteryl esters, ceramides and glycosylceramides, but not triglycerides, increased specifically in cells expressing the steatogenic HCV 3a core protein. Accordingly, inhibitors of cholesteryl ester biosynthesis such as statins and acyl-CoA cholesterol acyl transferase inhibitors prevented the increase of cholesteryl ester production and the formation of large lipid droplets in HCV core 3a-expressing cells. Furthermore, inhibition of de novo sphingolipid biosynthesis by myriocin - but not of glycosphingolipid biosynthesis by miglustat - affected both lipid droplet size and cholesteryl ester level. The lipid profile of purified lipid droplets, isolated from HCV 3a core-expressing cells, confirmed the particular increase of cholesteryl ester. Thus, both sphingolipid and cholesteryl ester biosynthesis are affected by the steatogenic core protein of HCV genotype 3a. These results may explain the peculiar lipid profile of HCV-infected patients with steatosis.
Collapse
Affiliation(s)
- Ursula Loizides-Mangold
- Department of Biochemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Sophie Clément
- Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
| | - Alba Alfonso-Garcia
- University of California Irvine, Beckman Laser Institute, Irvine, California, United States of America
| | - Emilie Branche
- Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
| | - Stéphanie Conzelmann
- Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
| | - Clotilde Parisot
- Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
| | - Eric O. Potma
- University of California Irvine, Beckman Laser Institute, Irvine, California, United States of America
| | - Howard Riezman
- Department of Biochemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Francesco Negro
- Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
- Divisions of Gastroenterology and Hepatology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
27
|
Martín-Vílchez S, Rodríguez-Muñoz Y, López-Rodríguez R, Hernández-Bartolomé Á, Borque-Iñurrita MJ, Molina-Jiménez F, García-Buey L, Moreno-Otero R, Sanz-Cameno P. Inhibition of tyrosine kinase receptor Tie2 reverts HCV-induced hepatic stellate cell activation. PLoS One 2014; 9:e106958. [PMID: 25302785 PMCID: PMC4193738 DOI: 10.1371/journal.pone.0106958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/12/2014] [Indexed: 01/18/2023] Open
Abstract
Background Hepatitis C virus (HCV) infection is a major cause of chronic liver disease (CLD) and is frequently linked to intrahepatic microvascular disorders. Activation of hepatic stellate cells (HSC) is a central event in liver damage, due to their contribution to hepatic renewal and to the development of fibrosis and hepatocarcinoma. During the progression of CLDs, HSC attempt to restore injured tissue by stimulating repair processes, such as fibrosis and angiogenesis. Because HSC express the key vascular receptor Tie2, among other angiogenic receptors and mediators, we analyzed its involvement in the development of CLD. Methods Tie2 expression was monitored in HSC cultures that were exposed to media from HCV-expressing cells (replicons). The effects of Tie2 blockade on HSC activation by either neutralizing antibody or specific signaling inhibitors were also examined. Results Media from HCV-replicons enhanced HSC activation and invasion and upregulated Tie2 expression. Notably, the blockade of Tie2 receptor (by a specific neutralizing antibody) or signaling (by selective AKT and MAPK inhibitors) significantly reduced alpha-smooth muscle actin (α-SMA) expression and the invasive potential of HCV-conditioned HSC. Conclusions These findings ascribe a novel profibrogenic function to Tie2 receptor in the progression of chronic hepatitis C, highlighting the significance of its dysregulation in the evolution of CLDs and its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Samuel Martín-Vílchez
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Yolanda Rodríguez-Muñoz
- Unidad de Hepatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-ehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rosario López-Rodríguez
- Unidad de Hepatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-ehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ángel Hernández-Bartolomé
- Unidad de Hepatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - María Jesús Borque-Iñurrita
- Unidad de Biología Molecular, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Francisca Molina-Jiménez
- Unidad de Biología Molecular, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Luisa García-Buey
- Unidad de Hepatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-ehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ricardo Moreno-Otero
- Unidad de Hepatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-ehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Paloma Sanz-Cameno
- Unidad de Hepatología, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-ehd), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- * E-mail:
| |
Collapse
|
28
|
Yao Z, Song X, Cao S, Liang W, Lu W, Yang L, Zhang Z, Wei L. Role of the exogenous HCV core protein in the interaction of human hepatocyte proliferation and macrophage sub-populations. PLoS One 2014; 9:e108278. [PMID: 25265479 PMCID: PMC4180735 DOI: 10.1371/journal.pone.0108278] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/19/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The core protein of hepatitis C virus (HCV) is found in the cytoplasm and nuclei of infected cells, including hepatocytes and other cells in the liver. The core protein could be secreted as well. Resident liver macrophages are dependent on the tissue micro-environment and external stimuli to differentiate M1 and M2 hypotypes with distinct functions, and increased expression of the nuclear transcription factor STAT3 was seen in M2-polarized macrophages. In contrast to proinflammatory M1 macrophages, M2 macrophages serve beneficial roles in chronic inflammation, immunosuppression, and tumorigenesis. METHODS Monocyte-derived human macrophage line (mTHP-1) was treated with the exogenous HCV core protein. Next, the mTHP-1 culture supernatant or cell pellets were added to culture media of normal human liver cell line (L02). RESULTS Only the culture supernatant stimulated L02 cells proliferation, which was associated with phosphorylated ERK expression. Core protein activated mTHP-1 cells showed enhanced pro- and anti-inflammatory cytokines secretion, which was accompanied by high expression of phosphorylated NF-κB105 and NF-κB65. However, phosphorylated STAT1, and STAT3, which are normally associated with M1 and M2 macrophage polarization, and cell surface expression of CD206, CD14, CD16, and CD86, were unaltered. A transwell co-culture system showed that only in mTHP-1 co-cultured with L02 in the presence of exogenous core protein, were higher levels of phosphorylated STAT3 and CD206 seen. CONCLUSIONS We showed L02 cells proliferation was accelerated by the culture supernatant of mTHP-1 cells treated with the exogenous HCV core protein. The exogenous core protein mediated the interaction between macrophages and hepatocytes in co-culture, which enhanced the expression of phosphorylated STAT3 and CD206 in macrophages.
Collapse
Affiliation(s)
- Zhiyan Yao
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Xiaotian Song
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Shiru Cao
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Wenzhang Liang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Wenran Lu
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Lijuan Yang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Zhengzheng Zhang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Lin Wei
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| |
Collapse
|
29
|
Renga B, Francisci D, Schiaroli E, Carino A, Cipriani S, D'Amore C, Sidoni A, Sordo RD, Ferri I, Lucattelli M, Lunghi B, Baldelli F, Fiorucci S. The HIV matrix protein p17 promotes the activation of human hepatic stellate cells through interactions with CXCR2 and Syndecan-2. PLoS One 2014; 9:e94798. [PMID: 24736615 PMCID: PMC3988079 DOI: 10.1371/journal.pone.0094798] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/19/2014] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The human immunodeficiency virus type 1 (HIV-1) p17 is a matrix protein involved in virus life's cycle. CXCR2 and Syndecan-2, the two major coreceptors for the p17 protein, are expressed in hepatic stellate cells (HSCs), a key cell type involved in matrix deposition in liver fibrotic disorders. AIM In this report we have investigated the in vitro impact of p17 on HSCs transdifferentiation and function and underlying signaling pathways involved in these processes. METHODS LX-2 cells, a human HSC line, and primary HSC were challenged with p17 and expressions of fibrogenic markers and of p17 receptors were assessed by qRT-PCR and Western blot. Downstream intracellular signaling pathways were evaluated with qRT-PCR and Western blot as well as after pre-treatment with specific pathway inhibitors. RESULTS Exposure of LX2 cells to p17 increases their contractile force, reshapes the cytoskeleton fibers and upregulates the expression of transdifferentiation markers including αSMA, COL1α1 and endothelin-1 through the activation of Jak/STAT and Rho signaling pathways. These effects are lost in HSCs pre-incubated with a serum from HIV positive person who underwent a vaccination with a p17 peptide. Confocal laser microscopy studies demonstrates that CXCR2 and syndecan-2 co-associate at the plasma membrane after exposure to p17. Immunostaining of HIV/HCV liver biopsies from co-infected patients reveals that the progression of liver fibrosis correlates with a reduced expression of CXCR2. CONCLUSIONS The HIV matrix protein p17 is pro-fibrogenic through its interactions both with CXCR2 and syndecan-2 on activated HSCs.
Collapse
Affiliation(s)
- Barbara Renga
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| | - Daniela Francisci
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Adriana Carino
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| | - Sabrina Cipriani
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Claudio D'Amore
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine and Biochemical Sciences, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy
| | - Rachele Del Sordo
- Department of Experimental Medicine and Biochemical Sciences, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy
| | - Ivana Ferri
- Department of Experimental Medicine and Biochemical Sciences, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy
| | | | | | - Franco Baldelli
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Fiorucci
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
30
|
Bose SK, Ray R. Hepatitis C virus infection and insulin resistance. World J Diabetes 2014; 5:52-58. [PMID: 24567801 PMCID: PMC3932427 DOI: 10.4239/wjd.v5.i1.52] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 12/20/2013] [Accepted: 01/14/2014] [Indexed: 02/05/2023] Open
Abstract
Approximately 170 million people worldwide are chronically infected with hepatitis C virus (HCV). Chronic HCV infection is the leading cause for the development of liver fibrosis, cirrhosis, hepatocellular carcinoma (HCC) and is the primary cause for liver transplantation in the western world. Insulin resistance is one of the pathological features in patients with HCV infection and often leads to development of type II diabetes. Insulin resistance plays an important role in the development of various complications associated with HCV infection. Recent evidence indicates that HCV associated insulin resistance may result in hepatic fibrosis, steatosis, HCC and resistance to anti-viral treatment. Thus, HCV associated insulin resistance is a therapeutic target at any stage of HCV infection. HCV modulates normal cellular gene expression and interferes with the insulin signaling pathway. Various mechanisms have been proposed in regard to HCV mediated insulin resistance, involving up regulation of inflammatory cytokines, like tumor necrosis factor-α, phosphorylation of insulin-receptor substrate-1, Akt, up-regulation of gluconeogenic genes like glucose 6 phosphatase, phosphoenolpyruvate carboxykinase 2, and accumulation of lipid droplets. In this review, we summarize the available information on how HCV infection interferes with insulin signaling pathways resulting in insulin resistance.
Collapse
|
31
|
Benzoubir N, Lejamtel C, Battaglia S, Testoni B, Benassi B, Gondeau C, Perrin-Cocon L, Desterke C, Thiers V, Samuel D, Levrero M, Bréchot C, Bourgeade MF. HCV core-mediated activation of latent TGF-β via thrombospondin drives the crosstalk between hepatocytes and stromal environment. J Hepatol 2013; 59:1160-8. [PMID: 23928402 DOI: 10.1016/j.jhep.2013.07.036] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 07/23/2013] [Accepted: 07/24/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS The mechanisms by which fibrosis, cirrhosis, and hepatocellular carcinoma (HCC) develop during chronic hepatitis C virus (HCV) infection are not fully understood. We previously observed that HCV core protein induced a TGF-β-dependent epithelial mesenchymal transition, a process contributing to the promotion of cell invasion and metastasis by impacting TGF-β1 signalling. Here we investigated HCV core capacity to drive increased expression of the active form of TGF-β1n transgenic mice and hepatoma cell lines. METHODS We used an in vivo model of HCV core expressing transgenic mice. RESULTS We observed that about 50% of genes deregulated by core protein expression were TGF-β1 target genes. Active TGF-β levels were increased in HCV core transgenic mouse livers. Overexpression of core protein in hepatoma cells increased active TGF-β levels in culture supernatants and induced Smad2/3 phosphorylation, thus reflecting activation of the TGF-β signaling pathway. Moreover, our data showed the implication of thrombospondin-1 in core-dependent TGF-β activation. Finally, hepatoma cells expressing HCV core could activate stellate cells in co-culture and this activation was TGF-β dependent. CONCLUSIONS Collectively, these data delineate a novel paradigm where HCV may be related to liver pathogenesis through its ability to induce a local, intrahepatic TGF-β activation. They argue for a dual impact of HCV core on liver fibrosis and liver carcinogenesis: HCV core could act both as autocrine and paracrine factor modulating TGF-β responses within hepatocytes and in stromal environment through TGF-β activation.
Collapse
Affiliation(s)
- Nassima Benzoubir
- INSERM U785, Villejuif F-94800, France; Univ Paris-Sud, UMR-S 785, Villejuif F-94800, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Nischalke HD, Berger C, Lutz P, Langhans B, Wolter F, Eisenhardt M, Krämer B, Kokordelis P, Glässner A, Müller T, Rosendahl J, Fischer J, Berg T, Grünhage F, Leifeld L, Soyka M, Nattermann J, Sauerbruch T, Stickel F, Spengler U. Influence of the CXCL1 rs4074 A allele on alcohol induced cirrhosis and HCC in patients of European descent. PLoS One 2013; 8:e80848. [PMID: 24260493 PMCID: PMC3832473 DOI: 10.1371/journal.pone.0080848] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/15/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND AIMS CXCL1 (CXC chemokine-ligand-1) is a ligand for CXC chemokine receptor 2 expressed on hepatic stellate cells (HSC). Thus, CXCL1 might contribute to HSC activation and fibrogenesis. In the present study, we investigated the influence of the CXCL1 rs4074 polymorphism on the occurrence of alcohol induced liver cirrhosis and hepatocellular carcinoma (HCC). METHODS The study involved 458 patients with alcoholic cirrhosis (170 with HCC), 115 alcoholics without liver disease and 342 healthy controls. All subjects were genotyped for the CXCL1 rs4074 polymorphism and CXCL1 serum levels of 132 patients were measured. In vitro CXCL1 secretion in TLR-transfected cell lines were studied by ELISA. RESULTS Distribution of the CXCL1 genotypes (GG/GA/AA) was 159/219/80 in patients with alcoholic cirrhosis, 52/44/19 in alcoholic controls and 158/140/44 in healthy controls. Patients with alcohol-induced cirrhosis were significantly more often carriers of the CXCL1 rs4074 A allele (65.3%) than alcoholics without liver disease (54.8%, OR=1.55; 95%CI=1.025-2.350; p=0.04) and healthy controls (53.8%, OR=1.62; 95%CI=1.212-2.151; p=0.001). Accordingly, the frequency of the CXCL1 rs4074 A allele was significantly higher in the cirrhotic patients than in the subjects without cirrhosis (41.4% vs. 33.9%, OR=1.38, 95% CI:1.14-1.66, p=0.001). Furthermore cirrhotic carriers of the CXCL1 rs4074 A allele had significantly higher CXCL1 serum levels than carriers of the GG genotype. In contrast to sera from healthy controls, sera from patients with alcoholic cirrhosis induced CXCL1 secretion in TLR2- (p=0.016) and TLR4- (p=0.008) transfected HEK293 cells. This finding indicates that sera from patients with alcoholic cirrhosis contain soluble ligands that can induce CXCL1 production via stimulation of TLRs. CONCLUSION The enhanced CXCL1 serum levels in carriers of the rs4074 A allele together with their increased frequency in patients with alcohol induced cirrhosis suggest the CXCL1 rs4074 A allele as a genetic risk factor for alcoholic cirrhosis.
Collapse
|
33
|
Rui W, Xie L, Liu X, He S, Wu C, Zhang X, Zhang L, Yang Y. Compound Astragalus and Salvia miltiorrhiza extract suppresses hepatocellular carcinoma progression by inhibiting fibrosis and PAI-1 mRNA transcription. JOURNAL OF ETHNOPHARMACOLOGY 2013; 151:198-209. [PMID: 24247078 DOI: 10.1016/j.jep.2013.10.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 09/26/2013] [Accepted: 10/05/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus membranaceus and Salvia miltiorrhiza have been used for centuries in China to treat liver diseases. Previous studies have shown that these herbs and their extracts inhibit the development of liver fibrosis and the proliferation and invasion of human hepatoma HepG2 cells. Further study of their pharmacological effects on hepatocellular carcinoma (HCC) is needed. To investigate the effects of Compound Astragalus and Salvia miltiorrhiza Extract (CASE) on diethylinitrosamine (DEN)-induced hepatocarcinogenesis in rats. MATERIALS AND METHODS Male rats were divided into five groups, with the first group serving as normal control, the second group receiving 0.2% DEN solution five times a week for 14 weeks, and the third to fifth group receiving the same DEN as in the second group together with CASE at the doses of 60, 120, and 240 mg/kg per day for 16 weeks, respectively. Hepatoma incidence, serum enzymes levels, degree of fibrosis and hydroxyproline content were evaluated and compared across the five groups to determine CASE's suppression of fibrosis and HCC progression. In addition, an in vitro experiment using HepG2 cells was conduct to verify CASE's effect on the transcription of plasminogen activator inhibitor-1 (PAI-1) mRNA. RESULTS CASE treatment significantly reduced the incidence and multiplicity of DEN-induced HCC development in a dose-dependent manner. It significantly suppressed the elevation of alanine transaminase, aspartate aminotransferase, gamma-glutamyl transferase, alkaline phosphatase, hyaluronic acid, direct bilirubin and total bilirubin, and significantly lessened the depression of serum total protein in DEN-induced HCC rats. CASE treatment also significantly suppressed the elevated expression of GST-P and α-SMA. The in vitro experiment confirmed that CASE inhibits the transcription of PAI-1 mRNA in HepG2 cells induced by TGF-β1 in a dose-dependent manner. CONCLUSIONS CASE suppresses DEN-induced hepatocarcinogenesis by inhibiting fibrosis and PAI-1 mRNA transcription, suggesting its potential clinical application in preventing and treating human HCC.
Collapse
Affiliation(s)
- Wenjuan Rui
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Lei Xie
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China; Department of Anesthesiology, Maternal and Child Care Hospital of Anhui Province, Hefei 230001, China
| | - Xin Liu
- Therapeutics Research Centre, University of Queensland, Princess Alexandra Hospital, Brisbane, Qld 4102, Australia
| | - Shufang He
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Chao Wu
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiaoxiang Zhang
- Department of Pharmaceutical Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Linjie Zhang
- Department of immunology, Anhui Medical University, Hefei 230032, China.
| | - Yan Yang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
34
|
Sun WY, Song Y, Hu SS, Wang QT, Wu HX, Chen JY, Wei W. Depletion of β-arrestin2 in hepatic stellate cells reduces cell proliferation via ERK pathway. J Cell Biochem 2013. [PMID: 23192415 DOI: 10.1002/jcb.24458] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
β-Arrestins are multifunctional adaptor proteins. Recently, some new roles of β-arrestins in regulating intracellular signaling networks have been discovered, which regulate cell growth, proliferation, and apoptosis. Though, the role of β-arrestins expression in the pathology of hepatic fibrosis remains unclear. In this study, the possible relationship between the expression of β-arrestins with the experimental hepatic fibrosis and the proliferation of hepatic stellate cells (HSCs) were investigated. Porcine serum induced liver fibrosis was established in this study. At five time points, the dynamic expression of β-arrestin1, β-arrestin2, and α-smooth muscle actin (α-SMA) in rat liver tissues, was measured by immunohistochemical staining, double immunofluorescent staining, and Western blotting. This study showed that aggravation of hepatic fibrosis with gradually increasing expression of β-arrestin2 in the hepatic tissues, but not β-arrestin1. Further, as hepatic fibrosis worsens, β-arrestin2-expressing activated HSCs accounts for an increasingly larger percentage of all activated HSCs. And the expression of β-arrestin2 had a significant positive correlation with the expression of α-SMA, an activated HSCs marker. In vitro studies, the dynamic expression of β-arrestin1 and β-arrestin2 in platelet derived growth factor-BB (PDGF-BB) stimulated HSCs was assessed by Western blotting. The expression of β-arrestin2 was remarkably increased in PDGF-BB stimulated HSCs. Furthermore, the small interfering RNA (siRNA) technique was used to explore the effect of β-arrestins on the proliferation of HSCs and the activation of ERK1/2. Transfection of siRNA targeting β-arrestin2 mRNA (siβ-arrestin2) into HSCs led to a 68% and 70% reduction of β-arrestin2 mRNA and protein expression, respectively. siβ-arrestin2 abolished the effect of PDGF-BB on the proliferation of HSCs. In addition, siβ-arrestin2 exerted the inhibition of the activation of ERK1/2 in HSCs. The present study provided strong evidence for the participation of the β-arrestin2 in the pathogenesis of hepatic fibrosis. The β-arrestin2 depletion diminishes HSCs ERK1/2 signaling and proliferation stimulated by PDGF-BB. Selective targeting of β-arrestin2 inhibitors to HSCs might present as a novel strategy for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Engineering Technology Research Center of Anti-inflammatory and Immunodrugs in Anhui Province, Hefei, Anhui Province 230032, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Clément S, Fauvelle C, Branche E, Kaddai V, Conzelmann S, Boldanova T, Bartosch B, Minehira K, Negro F. Role of seipin in lipid droplet morphology and hepatitis C virus life cycle. J Gen Virol 2013; 94:2208-2214. [PMID: 23907395 DOI: 10.1099/vir.0.054593-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Infectious hepatitis C virus (HCV) particle assembly starts at the surface of lipid droplets, cytoplasmic organelles responsible for neutral fat storage. We analysed the relationship between HCV and seipin, a protein involved in lipid droplet maturation. Although seipin overexpression did not affect the total mean volume occupied by lipid droplets nor the total triglyceride and cholesterol ester levels per cell, it caused an increase in the mean diameter of lipid droplets by 60 %, while decreasing their total number per cell. The latter two effects combined resulted in a 34 % reduction of the total outer surface area of lipid droplets per cell, with a proportional decrease in infectious viral particle production, probably due to a defect in particle assembly. These results suggest that the available outer surface of lipid droplets is a critical factor for HCV release, independent of the neutral lipid content of the cell.
Collapse
Affiliation(s)
- Sophie Clément
- Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
| | - Catherine Fauvelle
- Department of Immunology and Pathology, Faculty of Medicine, University of Geneva, Switzerland
| | - Emilie Branche
- Department of Immunology and Pathology, Faculty of Medicine, University of Geneva, Switzerland
| | - Vincent Kaddai
- Department of Immunology and Pathology, Faculty of Medicine, University of Geneva, Switzerland
| | - Stéphanie Conzelmann
- Department of Immunology and Pathology, Faculty of Medicine, University of Geneva, Switzerland
| | - Tujana Boldanova
- Department of Biomedicine, University Hospital Basel, Switzerland
| | - Birke Bartosch
- CRCL, INSERM U1052, CNRS 5286, University of Lyon, France
| | - Kaori Minehira
- Department of Physiology, University of Lausanne, Switzerland
| | - Francesco Negro
- Gastroenterology and Hepatology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland.,Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, Geneva, Switzerland
| |
Collapse
|
36
|
Langhans B, Krämer B, Louis M, Nischalke HD, Hüneburg R, Staratschek-Jox A, Odenthal M, Manekeller S, Schepke M, Kalff J, Fischer HP, Schultze JL, Spengler U. Intrahepatic IL-8 producing Foxp3⁺CD4⁺ regulatory T cells and fibrogenesis in chronic hepatitis C. J Hepatol 2013; 59:229-35. [PMID: 23624000 DOI: 10.1016/j.jhep.2013.04.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/04/2013] [Accepted: 04/07/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Regulatory CD4(+) T cells (Tregs) are considered to affect outcomes of HCV infection, because they increase in number during chronic hepatitis C and can suppress T-cell functions. METHODS Using microarray analysis, in situ immunofluorescence, ELISA, and flowcytometry, we characterised functional differentiation and localisation of adaptive Tregs in patients with chronic hepatitis C. RESULTS We found substantial upregulation of IL-8 in Foxp3(+)CD4(+) Tregs from chronic hepatitis C. Activated GARP-positive IL-8(+) Tregs were particularly enriched in livers of patients with chronic hepatitis C in close proximity to areas of fibrosis and their numbers were correlated with the stage of fibrosis. Moreover, Tregs induced upregulation of profibrogenic markers TIMP1, MMP2, TGF-beta1, alpha-SMA, collagen, and CCL2 in primary human hepatic stellate cells (HSC). HSC activation, but not Treg suppressor function, was blocked by adding a neutralizing IL-8 antibody. CONCLUSIONS Our studies identified Foxp3(+)CD4(+) Tregs as an additional intrahepatic source of IL-8 in chronic hepatitis C acting on HSC. Thus, Foxp3(+)CD4(+) Tregs in chronic hepatitis C have acquired differentiation as regulators of fibrogenesis in addition to suppressing local immune responses.
Collapse
Affiliation(s)
- Bettina Langhans
- Department of Internal Medicine I, University of Bonn, Bonn, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Dong ZX, Su L, Brymora J, Bird C, Xie Q, George J, Wang JH. Resistin mediates the hepatic stellate cell phenotype. World J Gastroenterol 2013; 19:4475-4485. [PMID: 23901222 PMCID: PMC3725371 DOI: 10.3748/wjg.v19.i28.4475] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 02/13/2013] [Accepted: 03/29/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To describe the role of resistin in liver fibrosis.
METHODS: For the in vivo animal study, Sprague Dawley rats were subjected to bile duct ligation (BDL) for 4 wk. Rat liver, adipose tissue (epididymal fat) and serum were analyzed for resistin expression. For the in vitro experiment, rat primary hepatic stellate cells (HSCs) and Kupffer cells (KCs) were used. HSCs were exposed to recombinant resistin, and collagen I, transforming growth factor β1, α smooth muscle actin, tissue inhibitor of metalloproteinase 1 and connective tissue growth factor expression were analyzed. Resistin gene and protein expression was quantified as was the expression of pro-inflammatory cytokines including tumor necrosis factor α (TNFα), interleukin (IL)-1, IL-6, IL-8 and monocyte chemotactic protein-1 (MCP-1). The effects of resistin on HSC proliferation, migration and apoptosis were determined. The effects of resistin on KCs were also investigated.
RESULTS: Following BDL, rat epididymal fat and serum rather than liver showed higher resistin expression compared to control rats. In liver, resistin was expressed in quiescent HSCs and KCs. Resistin treatment resulted in enhancement of TNFα, IL-6, IL-8 and MCP-1 gene expression and increased IL-6 and MCP-1 protein in HSCs. Resistin activated HSC phospho-MAPK/p38, and p38 inhibition diminished IL-6 and MCP-1 expression. Furthermore, resistin facilitated HSC proliferation and migration, but decreased apoptosis which was via an IL-6 and MCP-1 mechanism. Finally, resistin-induced transforming growth factor β1 from KCs enhanced HSC collagen Iexpression.
CONCLUSION: Resistin directly and indirectly modulates HSC behavior towards a more pro-fibrogenic phenotype.
Collapse
|
38
|
Wu CF, Lin YL, Huang YT. Hepatitis C virus core protein stimulates fibrogenesis in hepatic stellate cells involving the obese receptor. J Cell Biochem 2013; 114:541-50. [PMID: 22961938 DOI: 10.1002/jcb.24392] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 08/30/2012] [Indexed: 12/17/2022]
Abstract
Hepatitis C virus core protein (HCVcp), which is secreted by infected cells, is reported as an immunomodulator in immune cells. However, the effects of HCVcp on hepatic stellate cells (HSCs), the key cells in liver fibrosis, still remain unclear. In this study, we investigated the effects of HCVcp on obese receptor (ObR) related downstream signaling pathways and fibrogenic gene expression in HSCs. LX-2, a human HSC line, was incubated with HCVcp. Inhibitors and short interfering RNAs were used to interrogate the mechanisms of HCVcp action on HSCs. HCVcp (20-100 ng/ml) concentration-dependently stimulated α-smooth muscle actin (α-SMA) protein expression and mRNA expression of α-SMA, procollagen α2(I) and TGF-β1 genes, with a plateau of 220% of controls at 100 ng/ml. HCVcp induced mRNA and protein expression of ObR. Blocking of Ob-Rb with a neutralizing antibody inhibited phosphorylation of signal transducer and activator of transcription 3 (STAT3) and AMPKα stimulated by HCVcp. Furthermore, knockdown of Ob-Rb down-regulated HCVcp-induced STAT3, AKT, and AMPKα phosphorylation, and reversed HCVcp-suppressed mRNA expression of matrix metalloproteinase (MMP)-1, peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element binding protein-1c (SREBP-1c) genes. AMPKα signaling blockade reversed HCVcp-suppressed SREBP-1c mRNA expression. HCVcp stimulated reactive oxygen species formation and gp91(phox) (a component of NADPH oxidase) protein expression, together with AKT phosphorylation, leading to suppression of PPARγ and SREBP-1c genes. Our results provide a new finding that HCVcp induced ObR-dependent Janus Kinase (JAK) 2-STAT3, AMPKα, and AKT signaling pathways and modulated downstream fibrogenetic gene expression in HSCs.
Collapse
Affiliation(s)
- Ching-Fen Wu
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | |
Collapse
|
39
|
Hepatitis C virus core protein epigenetically silences SFRP1 and enhances HCC aggressiveness by inducing epithelial-mesenchymal transition. Oncogene 2013; 33:2826-35. [PMID: 23770846 DOI: 10.1038/onc.2013.225] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most deadly cancers. Aberrant oncogenic activation of the Wnt/β-catenin signaling pathway contributes to hepatocellular carcinogenesis. Various epigenetic modifications of the Wnt antagonist secreted frizzled-related protein (SFRP) family have been implicated in regulating Wnt signaling. Here, we report that Hepatitis C virus (HCV) core protein downregulates SFRP1 expression when it is expressed in Huh7 and HepG2 cells. SFRP1 expression can be effectively restored by using either a DNA methylation inhibitor alone or in combination with a histone deacetylase inhibitor. DNA methylation analysis of the SFRP1 promoter revealed that cytosine-phosphate-guanine (CpG) islands close to the transcriptional start site (TSS) in the SFRP1 promoter were hypermethylated in core-expressing Huh7 cells, suggesting that HCV core protein may downregulate SFRP1 expression by inducing hypermethylation of the SFRP1 promoter. Chromatin immunoprecipitation revealed that HCV core protein markedly increased the expression level and binding of DNA methyltransferase-1 (Dnmt1) and histone deacetylase-1 (HDAC1) to the TSS of the SFRP1 promoter region, resulting in repression of acetyl-histone H3-binding capacity to SFRP1 promoter and the eventual epigenetic silencing of SFRP1 expression. Furthermore, the core protein-promoted cell proliferation, migration and invasiveness were effectively abrogated either by Dnmt1 knockdown or restoration of SFRP1 expression in hepatoma cells. Dnmt1 knockdown or SFRP1 overexpression also inhibited HCV core-induced epithelial-mesenchymal transition (EMT) and significantly decreased the expression levels of activated β-catenin and Wnt/β-catenin target genes, c-Myc and cyclin D1. We further showed that knockdown of Dnmt1 and restoration of SFRP1 inhibited core-induced in vivo tumor growth and aggressiveness in a xenograft HCC model. Taken together, our results strongly suggest that the HCV core-induced epigenetic silencing of SFRP1 may lead to the activation of the Wnt signaling pathway and thus contribute to HCC aggressiveness through induction of EMT.
Collapse
|
40
|
Zan Y, Zhang Y, Tien P. Hepatitis B virus e antigen induces activation of rat hepatic stellate cells. Biochem Biophys Res Commun 2013; 435:391-6. [PMID: 23665329 DOI: 10.1016/j.bbrc.2013.04.098] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 04/29/2013] [Indexed: 01/11/2023]
Abstract
Chronic hepatitis B virus infection is a major cause of hepatic fibrosis, leading to liver cirrhosis and hepatocellular carcinoma. Hepatitis B virus e antigen (HBeAg) is an accessory protein of HBV, not required for viral replication but important for natural infection in vivo. Hepatic stellate cells (HSCs) are the major producers of excessive extracellular matrix during liver fibrogenesis. Therefore, we examined the influence of HBeAg on HSCs. The rat HSC line HSC-T6 was transfected with HBeAg plasmids, and expression of α-smooth muscle actin, collagen I, transforming growth factor-β1 (TGF-β), and tissue inhibitors of metalloproteinase 1 (TIMP-1) was investigated by quantitative real-time PCR. The proliferation of HSCs was determined by MTS analysis. HBeAg transduction induced up-regulation of these fibrogenic genes and proliferation of HSCs. We found that HBeAg induced TGF-β secretion in HSCs, and the activation of HSCs was prevented by a neutralizing anti-TGF-β antibody. Depletion and addition of HBeAg protein in conditioned medium from HSC-T6 cells transduced with HBeAg indicated that HBeAg directly induced the activation and proliferation of rat primary HSCs. Taken together, HBeAg induces the activation and proliferation of HSCs, mainly mediated by TGF-β, and HBeAg protein purified from cell medium can directly activate HSCs.
Collapse
Affiliation(s)
- Yanlu Zan
- Center for Molecular Virology, CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
| | | | | |
Collapse
|
41
|
Bai Q, An J, Wu X, You H, Ma H, Liu T, Gao N, Jia J. HBV promotes the proliferation of hepatic stellate cells via the PDGF-B/PDGFR-β signaling pathway in vitro. Int J Mol Med 2012; 30:1443-50. [PMID: 23042547 DOI: 10.3892/ijmm.2012.1148] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 07/10/2012] [Indexed: 12/25/2022] Open
Abstract
The activation of hepatic stellate cells (HSCs) is closely associated with liver fibrosis in chronic hepatitis B virus (HBV) infection. However, the molecular mechanisms leading to HSC activation remain unclear. It has been reported that the platelet-derived growth factor-B (PDGF-B)/PDGF receptor-β (PDGFR-β) signaling pathway is involved in this process. Thus, we investigated whether HBV and its protein contribute to HSC proliferation by the PDGF-B/PDGFR-β signaling pathway. HBV particles were purified from the supernatant of HepG2.2.15 cells by ultracentrifugation and the cell lines carrying HBV preS, e, c or x genes were obtained. After incubation with HBV particles or co-cultured with the cell lines expressed in the viral protein, the proliferation of LX-2 cells, an HSC cell line, were detected by flow cyto-metry and real-time PCR and the expression of molecules related to the PDGF-B/PDGFR-β signaling pathway were further measured. Our results indicated that HBV particles, c and x proteins promoted LX-2 proliferation and increased the mRNA levels of PDGF-B, PDGFR-β, collagen-I and α-smooth muscle actin (α-SMA), as well as the phosphorylation of PDGFR-β; however, the expression protein levels of PDGF-B and PDGFR-β remained unchanged. In conclusion, HBV particles and HBV c and x proteins promote HSC proliferation and fibrogenesis in vitro and the PDGF-B/PDGFR-β signaling pathway is important in this process.
Collapse
Affiliation(s)
- Qixuan Bai
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Alisi A, Arciello M, Petrini S, Conti B, Missale G, Balsano C. Focal adhesion kinase (FAK) mediates the induction of pro-oncogenic and fibrogenic phenotypes in hepatitis C virus (HCV)-infected cells. PLoS One 2012; 7:e44147. [PMID: 22937161 PMCID: PMC3429423 DOI: 10.1371/journal.pone.0044147] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 08/01/2012] [Indexed: 01/18/2023] Open
Abstract
Hepatitis C Virus (HCV) infection is one of the most common etiological factors involved in fibrosis development and its progression to hepatocellular carcinoma (HCC). The pivotal role of hepatic stellate cells (HCSs) and extracellular matrix (ECM) in fibrogenesis is now certainly accepted, while the network of molecular interactions connecting HCV is emerging as a master regulator of several biological processes including proliferation, inflammation, cytoskeleton and ECM remodeling. In this study, the effects of HCV proteins expression on liver cancer cells, both pro-invasive and pro-fibrogenic phenotypes were explored. As a model of HCV infection, we used permissive Huh7.5.1 hepatoma cells infected with JFH1-derived ccHCV. Conditioned medium from these cells was used to stimulate LX-2 cells, a line of HSCs. We found that the HCV infection of Huh7.5.1 cells decreased adhesion, increased migration and caused the delocalization of alpha-actinin from plasma membrane to cytoplasm and increased expression levels of paxillin. The treatment of LX-2 cells, with conditioned medium from HCV-infected Huh7.5.1 cells, caused an increase in cell proliferation, expression of alpha-smooth muscle actin, hyaluronic acid release and apoptosis rate measured as cleaved poly ADP-ribose polymerase (PARP). These effects were accompanied in Huh7.5.1 cells by an HCV-dependent increasing of FAK activation that physically interacts with phosphorylated paxillin and alpha-actinin, and a rising of tumor necrosis factor alpha production/release. Silencing of FAK by siRNA reverted all effects of HCV infection, both those directed on Huh7.5.1 cells, and those indirect effects on the LX-2 cells. Moreover and interestingly, FAK inhibition enhances apoptosis in HCV-conditioned LX-2 cells. In conclusion, our findings demonstrate that HCV, through FAK activation, may promote cytoskeletal reorganization and a pro-oncogenic phenotype in hepatocyte-like cells, and a fibrogenic phenotype in HSCs.
Collapse
Affiliation(s)
- Anna Alisi
- Liver Research Unit, Confocal Microscopy Facility of “Bambino Gesù” Children’s Hospital, IRCCS, Rome, Italy
- * E-mail: (AA); (CB)
| | - Mario Arciello
- Laboratory of Molecular Virology and Oncology, A. Cesalpino Foundation, University of Rome “La Sapienza”, Rome, Italy
- Department of Internal Medicine, University of L’Aquila, L’Aquila, Italy
| | - Stefania Petrini
- Confocal Microscopy Facility of “Bambino Gesù” Children’s Hospital, IRCCS, Rome, Italy
| | - Beatrice Conti
- Laboratory of Molecular Virology and Oncology, A. Cesalpino Foundation, University of Rome “La Sapienza”, Rome, Italy
- Department of Internal Medicine, University of L’Aquila, L’Aquila, Italy
| | - Gabriele Missale
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero – Universitaria di Parma, Parma, Italy
| | - Clara Balsano
- Laboratory of Molecular Virology and Oncology, A. Cesalpino Foundation, University of Rome “La Sapienza”, Rome, Italy
- IBPM (Institute of Biology and Molecular Pathology), Rome, Italy
- * E-mail: (AA); (CB)
| |
Collapse
|
43
|
Profibrogenic chemokines and viral evolution predict rapid progression of hepatitis C to cirrhosis. Proc Natl Acad Sci U S A 2012; 109:14562-7. [PMID: 22829669 DOI: 10.1073/pnas.1210592109] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chronic hepatitis C may follow a mild and stable disease course or progress rapidly to cirrhosis and liver-related death. The mechanisms underlying the different rates of disease progression are unknown. Using serial, prospectively collected samples from cases of transfusion-associated hepatitis C, we identified outcome-specific features that predict long-term disease severity. Slowly progressing disease correlated with an early alanine aminotransferase peak and antibody seroconversion, transient control of viremia, and significant induction of IFN-γ and MIP-1β, all indicative of an effective, albeit insufficient, adaptive immune response. By contrast, rapidly progressive disease correlated with persistent and significant elevations of alanine aminotransferase and the profibrogenic chemokine MCP-1 (CCL-2), greater viral diversity and divergence, and a higher rate of synonymous substitution. This study suggests that the long-term course of chronic hepatitis C is determined early in infection and that disease severity is predicted by the evolutionary dynamics of hepatitis C virus and the level of MCP-1, a chemokine that appears critical to the induction of progressive fibrogenesis and, ultimately, the ominous complications of cirrhosis.
Collapse
|
44
|
Nischalke HD, Berger C, Luda C, Müller T, Berg T, Coenen M, Krämer B, Körner C, Trebicka J, Grünhage F, Lammert F, Nattermann J, Sauerbruch T, Spengler U. The CXCL1 rs4074 A allele is associated with enhanced CXCL1 responses to TLR2 ligands and predisposes to cirrhosis in HCV genotype 1-infected Caucasian patients. J Hepatol 2012; 56:758-64. [PMID: 22173151 DOI: 10.1016/j.jhep.2011.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 10/11/2011] [Accepted: 10/12/2011] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS CXCL1 is a ligand for CXC chemokine-receptor 2 expressed on hepatic stellate cells (HSC). Thus, CXCL1 might contribute to HSC activation and fibrogenesis. Here, we investigated whether the CXCL1 rs4074 polymorphism affects CXCL1 expression and progression of chronic hepatitis C virus (HCV) infection towards cirrhosis. METHODS The study involved 237 patients with chronic HCV genotype 1 infection (75 with cirrhosis) and 342 healthy controls. The CXCL1 rs4074 polymorphism was determined by a LightSNiP assay on the LightCycler system. CXCL1 serum levels and induction in response to HCV proteins were studied by ELISA. RESULTS Distributions of CXCL1 genotypes (GG/GA/AA) matched the Hardy-Weinberg equilibrium in all subgroups (HCV-associated cirrhosis: 29.3%/54.7%/16.0%; non-cirrhotic HCV infection: 45.1%/44.4%/10.5%, healthy controls: 46.2%/40.9%/12.9%). HCV-infected cirrhotic patients had a significantly greater CXCL1 rs4074 A allele frequency (43.3%) than patients without cirrhosis (32.7%, OR=1.573, p=0.03) and healthy controls (33.3%, OR=1.529, p=0.02). In vitro carriers of the A allele produced greater amounts of CXCL1 in response to TLR2-ligands including HCV core and NS3, and HCV-infected carriers of the CXCL1 rs4074 A allele had higher CXCL1 serum levels than those with the G/G genotype. Moreover, multivariate Cox-regression analysis confirmed age and the presence of a CXCL1 rs4074 A allele as risk factors for cirrhosis. CONCLUSIONS Enhanced production of CXCL1 in response to HCV antigens in carriers of the rs4074 A allele together with its increased frequency in cirrhotic patients with hepatitis C suggest the CXCL1 rs4074 A allele as a genetic risk factor for cirrhosis progression in hepatitis C.
Collapse
Affiliation(s)
- Hans Dieter Nischalke
- Department of Internal Medicine I, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bellini A, Marini MA, Bianchetti L, Barczyk M, Schmidt M, Mattoli S. Interleukin (IL)-4, IL-13, and IL-17A differentially affect the profibrotic and proinflammatory functions of fibrocytes from asthmatic patients. Mucosal Immunol 2012; 5:140-9. [PMID: 22189956 DOI: 10.1038/mi.2011.60] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Fibrocytes contribute to the fibrotic changes most frequently observed in forms of asthma where inflammation is driven by T helper type 2 (Th2) cells. The mechanisms that regulate the profibrotic function of asthmatic fibrocytes are largely unknown. We isolated circulating fibrocytes from patients with allergen-exacerbated asthma, who showed the presence of fibrocytes, together with elevated concentrations of interleukin (IL)-4 and IL-13 and slightly increased concentrations of the Th17 cell-derived IL-17A, in induced sputum. Fibrocytes stimulated with IL-4 and IL-13 produced high levels of collagenous and non-collagenous matrix components and low levels of proinflammatory cytokines. Conversely, fibrocytes stimulated with IL-17A proliferated and released proinflammatory factors that may promote neutrophil recruitment and airway hyperresponsiveness. IL-17A also indirectly increased α-smooth muscle actin but not collagen expression in fibrocytes. Thus, fibrocytes may proliferate and express a predominant profibrotic or proinflammatory phenotype in asthmatic airways depending on the local concentrations of Th2- and Th17-derived cytokines.
Collapse
Affiliation(s)
- A Bellini
- Avail Biomedical Research Institute, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
46
|
Lee CM, Yen YH, Hung CH, Lu SN, Wang JH, Wang JC, Chen CH, Kee KM, Hu TH, Changchien CS. Liver interleukin-8 messenger RNA expression and interferon sensitivity-determining region mutations relate to treatment response in hepatitis C 1b. Antivir Ther 2012; 16:825-32. [PMID: 21900714 DOI: 10.3851/imp1868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND In vitro study has shown that mechanisms for inhibiting interferon (IFN)-α antiviral action by non-structural 5A protein include interaction with IFN-induced RNA-dependent protein kinase and induction of interleukin (IL)-8 expression. Mutations in the non-structural 5A IFN sensitivity-determining region (ISDR) were reported to correlate with sustained virological response (SVR). IL-8 is associated with the inhibition of IFN-α action. We investigated whether pretreatment ISDR mutations and hepatic IL-8 messenger RNA (mRNA) expression had an effect on the SVR rate under combination therapy. METHODS A total of 53 HCV-1b patients who completed 24 weeks of pegylated-IFN-α2b plus ribavirin, a 24-week follow-up and had enough tissue specimens were enrolled. Liver biopsy was performed within 6 months before antiviral therapy. Hepatic IL-8 mRNA expression was measured by real-time reverse transcriptase PCR. RESULTS Of 53 patients, 30 exhibited SVR. Multivariate analysis revealed that hepatic IL-8 mRNA expression <1.5×10(-4) (OR 6.66, 95% CI 1.77-25.05) and ISDR mutations ≥4 (OR 12.20, 95% CI 1.23-125.00) were independent predictors of SVR. Fibrosis scores and alanine aminotransferase levels were predictive of hepatic IL-8 mRNA expression by multiple linear regression analysis (r(2)=0.204). CONCLUSIONS SVR to combination therapy in hepatitis C 1b patients was associated with down-regulated hepatic IL-8 mRNA expression and ISDR mutations. Fibrosis scores and alanine aminotransferase levels were predictive of hepatic IL-8 mRNA expression.
Collapse
Affiliation(s)
- Chuan-Mo Lee
- Division of Hepatogastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Clément S, Peyrou M, Sanchez-Pareja A, Bourgoin L, Ramadori P, Suter D, Vinciguerra M, Guilloux K, Pascarella S, Rubbia-Brandt L, Negro F, Foti M. Down-regulation of phosphatase and tensin homolog by hepatitis C virus core 3a in hepatocytes triggers the formation of large lipid droplets. Hepatology 2011; 54:38-49. [PMID: 21465511 DOI: 10.1002/hep.24340] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Hepatitis C virus (HCV) perturbs the host's lipid metabolism and often results in hepatic steatosis. In nonalcoholic fatty liver disease, the intrahepatic down-regulation of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a critical mechanism leading to steatosis and its progression toward fibrosis and hepatocellular carcinoma. However, whether an HCV infection triggers the formation of large lipid droplets through PTEN-dependent mechanisms is unknown. We assessed PTEN expression in the livers of patients infected with HCV genotype 1 or 3 with or without steatosis. The role of PTEN in the HCV-induced biogenesis of lipid droplets was further investigated in vitro with hepatoma cells transduced with the HCV core protein of genotype 1b or 3a. Our data indicate that PTEN expression was down-regulated at the posttranscriptional level in steatotic patients infected with genotype 3a. Similarly, the in vitro expression of the HCV genotype 3a core protein (but not 1b), typically leading to the appearance of large lipid droplets, down-regulated PTEN expression by a mechanism involving a microRNA-dependent blockade of PTEN messenger RNA translation. PTEN down-regulation promoted in turn a reduction of insulin receptor substrate 1 (IRS1) expression. Interestingly, either PTEN or IRS1 overexpression prevented the development of large lipid droplets, and this indicates that the down-regulation of both PTEN and IRS1 is required to affect the biogenesis of lipid droplets. However, IRS1 knockdown per se did not alter the morphology of lipid droplets, and this suggests that other PTEN-dependent mechanisms are involved in this process. CONCLUSION The down-regulation of PTEN and IRS1 is a critical event leading to the HCV genotype 3a-induced formation of large lipid droplets in hepatocytes.
Collapse
Affiliation(s)
- Sophie Clément
- Division of Clinical Pathology, University Hospital, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Zimmermann HW, Seidler S, Gassler N, Nattermann J, Luedde T, Trautwein C, Tacke F. Interleukin-8 is activated in patients with chronic liver diseases and associated with hepatic macrophage accumulation in human liver fibrosis. PLoS One 2011; 6:e21381. [PMID: 21731723 PMCID: PMC3120868 DOI: 10.1371/journal.pone.0021381] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/29/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Interleukin-8 (IL-8, CXCL8) is a potent chemoattractant for neutrophils and contributes to acute liver inflammation. Much less is known about IL-8 in chronic liver diseases (CLD), but elevated levels were reported from alcoholic and hepatitis C-related CLD. We investigated the regulation of IL-8, its receptors CXCR1 and CXCR2 and possible IL-8 responding cells in CLD patients. METHODOLOGY Serum IL-8 levels were measured in CLD patients (n = 200) and healthy controls (n = 141). Intrahepatic IL-8, CXCR1 and CXCR2 gene expression was quantified from liver samples (n = 41), alongside immunohistochemical neutrophil (MPO) and macrophage (CD68) stainings. CXCR1 and CXCR2 expression was analyzed on purified monocytes from patients (n = 111) and controls (n = 31). In vitro analyses explored IL-8 secretion by different leukocyte subsets. PRINCIPAL FINDINGS IL-8 serum levels were significantly increased in CLD patients, especially in end-stage cirrhosis. Interestingly, patients with cholestatic diseases exhibited highest IL-8 serum concentrations. IL-8 correlated with liver function, inflammatory cytokines and non-invasive fibrosis markers. Intrahepatically, IL-8 and CXCR1 expression were strongly up-regulated. However, intrahepatic IL-8 could only be associated to neutrophil infiltration in patients with primary biliary cirrhosis (PBC). In non-cholestatic cirrhosis, increased IL-8 and CXCR1 levels were associated with hepatic macrophage accumulation. In line, CXCR1, but not CXCR2 or CXCR3, expression was increased on circulating monocytes from cirrhotic patients. Moreover, monocyte-derived macrophages from CLD patients, especially the non-classical CD16⁺ subtype, displayed enhanced IL-8 secretion in vitro. CONCLUSIONS IL-8 is strongly activated in CLD, thus likely contributing to hepatic inflammation. Our study suggests a novel role of IL-8 for recruitment and activation of hepatic macrophages via CXCR1 in human liver cirrhosis.
Collapse
Affiliation(s)
| | | | | | | | - Tom Luedde
- Department of Medicine III, University Hospital, Aachen, Germany
| | | | - Frank Tacke
- Department of Medicine III, University Hospital, Aachen, Germany
| |
Collapse
|
49
|
Pascarella S, Clément S, Guilloux K, Conzelmann S, Penin F, Negro F. Effects of hepatitis C virus on suppressor of cytokine signaling mRNA levels: Comparison between different genotypes and core protein sequence analysis. J Med Virol 2011; 83:1005-15. [DOI: 10.1002/jmv.22072] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
50
|
Abstract
It is evidenced that both HBV and HCV infections may cause chronic hepatitis, liver cirrhosis and hepatocellular carcinoma (HCC). HCV is considered as an oncogen virus. The mechanism of carcinogenesis in case of the two distinct viruses shows a number of common and different features. HCC usually develops in stage of liver cirrhosis caused by chronic inflammation of many years or decades. In contrast to HBC, HCV will not be incorporated into the genome of hepatocytes. It induces throughout different mechanisms the development of cancer cells, which will proliferate out of control. Before the discovery of HCV, the connection of non-A non-B (NANB) hepatitis and liver cancer has yet been observed. The subsequent epidemiological studies confirmed this association. A number of publications prove that HCC may develop without cirrhosis in HCV infected patients. The exact mechanism is not known, but data indicate that some proteins of the virus may induce oncogenic process. Differences were found in HCV core gene between HCC patients with and without cirrhosis. It is postulated that the virus evolves its oncogenic effect via endoplasmic and oxidative stress, further on by activation of different oncogenic signal pathways. Recent publications indicate the role of virus induced insulin resistance. Besides the virus determined factors, the host reaction, the deficient tumor cell eliminating capacity may also have a role in the development of liver carcinoma. Monitoring of HCV infected patients for HCC is an important clinical issue. Most relevant is the regular ultrasound cheek up. Serum alpha fetoprotein level is elevated only in a smaller proportion of the patients. The treatment protocol of HCV induced HCC does not differ from the others developed in non-HCV liver diseases.
Collapse
Affiliation(s)
- Ferenc Szalay
- Semmelweis Egyetem, Altalános Orvostudományi Kar, I. Belgyógyászati Klinika, Budapest.
| |
Collapse
|