1
|
Malekshahi A, Alamdary A, Safarzadeh A, Khavandegar A, Nikoo HR, Safavi M, Ajorloo M, Bahavar A, Ajorloo M. Potential roles of core and core+1 proteins during the chronic phase of hepatitis C virus infection. Future Virol 2023. [DOI: 10.2217/fvl-2022-0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
The HCV Core protein is a multifunctional protein that interacts with many viral and cellular proteins. In addition to the encapsidation of the viral genome, it can disturb various cellular pathways and impede antiviral cellular responses such as interferon (IFN) production. The Core protein can also disrupt the functions of immune cells against HCV. The Core protein helps viral infection persistency by interfering with apoptosis. The Core+1 protein plays a significant role in inducing chronic HCV infection through diverse mechanisms. We review some of the mechanisms by which Core and Core+1 proteins facilitate HCV infection to chronic infection. These proteins could be considered for designing more sufficient treatments and effective vaccines against HCV.
Collapse
Affiliation(s)
- Asra Malekshahi
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ashkan Alamdary
- Department of Biology, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Safarzadeh
- Department of Biology, University of Padova, Padova, Italy
| | - Armin Khavandegar
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Hadi Razavi Nikoo
- Infectious Disease Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahshid Safavi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Mobina Ajorloo
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Atefeh Bahavar
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Ajorloo
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
2
|
Nakayama J, Gong Z. Transgenic zebrafish for modeling hepatocellular carcinoma. MedComm (Beijing) 2020; 1:140-156. [PMID: 34766114 PMCID: PMC8491243 DOI: 10.1002/mco2.29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Liver cancer is the third leading cause of cancer‐related deaths throughout the world, and more than 0.6 million people die from liver cancer annually. Therefore, novel therapeutic strategies to eliminate malignant cells from liver cancer patients are urgently needed. Recent advances in high‐throughput genomic technologies have identified de novo candidates for oncogenes and pharmacological targets. However, testing and understanding the mechanism of oncogenic transformation as well as probing the kinetics and therapeutic responses of spontaneous tumors in an intact microenvironment require in vivo examination using genetically modified animal models. The zebrafish (Danio rerio) has attracted increasing attention as a new model for studying cancer biology since the organs in the model are strikingly similar to human organs and the model can be genetically modified in a short time and at a low cost. This review summarizes the current knowledge of epidemiological data and genetic alterations in hepatocellular carcinoma (HCC), zebrafish models of HCC, and potential therapeutic strategies for targeting HCC based on knowledge from the models.
Collapse
Affiliation(s)
- Joji Nakayama
- Department of Biological Sciences National University of Singapore Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences National University of Singapore Singapore
| |
Collapse
|
3
|
Ferrari C, Barili V, Varchetta S, Mondelli MU. Immune Mechanisms of Viral Clearance and Disease Pathogenesis During Viral Hepatitis. THE LIVER 2020:821-850. [DOI: 10.1002/9781119436812.ch63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Navas MC, Stoll-Keller F, Pavlovic J. Lack of expression of hepatitis C virus core protein in human monocyte-erived dendritic cells using recombinant semliki forest virus. ACTA BIOLÓGICA COLOMBIANA 2019. [DOI: 10.15446/abc.v24n3.79368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C Virus belongs to the Flaviviridae family. One proposed mechanism of HCV persistence in the ability to infect hematopoietic cells, including Dendritic cells (DCs). HCV infection of DCs could impair their functions that represent one of the mechanisms, thus hampering viral clearance by the host immune system. Among HCV-encoded proteins, the highly conserved Core protein has been suggested to be responsible for the immunomodulatory properties of this Hepacivirus. Recombinant viral vectors expressing the HCV Core protein and allowing its transduction and therefore the expression of the protein into DCs could be useful tools for the analysis of the properties of the Core protein. Vaccinia Virus and retrovirus have been used to transduce human DCs. Likewise, gene transfer into DCs using Semliki Forest Virus has been reported. This study aimed to express the HCV Core protein in human monocyte-derived DCs using an SFV vector, in which the subgenomic RNA encoding the structural proteins was replaced by the HCV Core sequence and then analyze the effects of its expression on DCs functions.
Collapse
|
5
|
Feige MH, Vieth M, Sokolova O, Täger C, Naumann M. Helicobacter pylori induces direct activation of the lymphotoxin beta receptor and non-canonical nuclear factor-kappa B signaling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:545-550. [PMID: 29329668 DOI: 10.1016/j.bbamcr.2018.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/21/2017] [Accepted: 01/08/2018] [Indexed: 01/25/2023]
Abstract
The pathogen Helicobacter pylori, which infects half of the world's population, is a major risk factor for the development of gastric diseases including chronic gastritis and gastric cancer. Among H. pylori's virulence factors is the cytotoxin-associated gene pathogenicity island (cagPAI), which encodes for a type IV secretion system (T4SS). The T4SS induces fast canonical nuclear factor-kappa B (NF-κB) signaling, a major factor increasing inflammation, supressing apoptotic cell death and thereby promoting the development of neoplasia. However, H. pylori's capability to mediate fast non-canonical NF-κB signaling is unresolved, despite a contribution of non-canonical NF-κB signaling to gastric cancer has been suggested. We analyzed signaling elements within non-canonical NF-κB in response to H. pylori in epithelial cell lines by immunoprecipitation, immunoblot, electrophoretic mobility shift assay and RNA interference knockdown. In addition, tissue samples of H. pylori-infected patients were investigated by immunohistochemistry. Here, we provide evidence for a T4SS-dependent direct activation of non-canonical NF-κB signaling. We identified the lymphotoxin beta receptor (LTβR) to elicit the fast release of NF-κB inducing kinase (NIK) from the receptor complex leading to non-canonical NF-κB signaling. Further, NIK expression was increased in human biopsies of H. pylori-associated gastritis. Thus, NIK could represent a novel target to reduce Helicobacter pylori-induced gastric inflammation and pathology.
Collapse
Affiliation(s)
- Michael Hartmut Feige
- Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Olga Sokolova
- Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Christian Täger
- Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany.
| |
Collapse
|
6
|
Koroleva EP, Fu YX, Tumanov AV. Lymphotoxin in physiology of lymphoid tissues - Implication for antiviral defense. Cytokine 2016; 101:39-47. [PMID: 27623349 DOI: 10.1016/j.cyto.2016.08.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022]
Abstract
Lymphotoxin (LT) is a member of the tumor necrosis factor (TNF) superfamily of cytokines which serves multiple functions, including the control of lymphoid organ development and maintenance, as well as regulation of inflammation and autoimmunity. Although the role of LT in organogenesis and maintenance of lymphoid organs is well established, the contribution of LT pathway to homeostasis of lymphoid organs during the immune response to pathogens is less understood. In this review, we highlight recent advances on the role of LT pathway in antiviral immune responses. We discuss the role of LT signaling in lymphoid organ integrity, type I IFN production and regulation of protection and immunopathology during viral infections. We further discuss the potential of therapeutic targeting LT pathway for controlling immunopathology and antiviral protection.
Collapse
Affiliation(s)
- Ekaterina P Koroleva
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas School of Medicine, UT Health Science Center, San Antonio, TX, USA; Trudeau Institute, Saranac Lake, NY
| | - Yang-Xin Fu
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexei V Tumanov
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas School of Medicine, UT Health Science Center, San Antonio, TX, USA; Trudeau Institute, Saranac Lake, NY.
| |
Collapse
|
7
|
Vegna S, Gregoire D, Moreau M, Lassus P, Durantel D, Assenat E, Hibner U, Simonin Y. NOD1 Participates in the Innate Immune Response Triggered by Hepatitis C Virus Polymerase. J Virol 2016; 90:6022-6035. [PMID: 27099311 PMCID: PMC4907226 DOI: 10.1128/jvi.03230-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/13/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Hepatitis C virus (HCV) triggers innate immunity signaling in the infected cell. Replication of the viral genome is dispensable for this phenotype, and we along with others have recently shown that NS5B, the viral RNA-dependent RNA polymerase, synthesizes double-stranded RNA (dsRNA) from cellular templates, thus eliciting an inflammatory response, notably via activation of type I interferon and lymphotoxin β. Here, we investigated intracellular signal transduction pathways involved in this process. Using HepaRG cells, a model that largely recapitulates the in vivo complexities of the innate immunity receptor signaling, we have confirmed that NS5B triggered increased expression of the canonical pattern recognition receptors (PRRs) specific for dsRNA, namely, RIG-I, MDA5, and Toll-like receptor 3 (TLR3). Unexpectedly, intracellular dsRNA also led to accumulation of NOD1, a receptor classically involved in recognition of bacterial peptidoglycans. NOD1 activation, confirmed by analysis of its downstream targets, was likely due to its interaction with dsRNA and was independent of RIG-I and mitochondrial antiviral signaling protein (MAVS/IPS-1/Cardif/VISA) signaling. It is likely to have a functional significance in the cellular response in the context of HCV infection since interference with the NOD1 pathway severely reduced the inflammatory response elicited by NS5B. IMPORTANCE In this study, we show that NOD1, a PRR that normally senses bacterial peptidoglycans, is activated by HCV viral polymerase, probably through an interaction with dsRNA, suggesting that NOD1 acts as an RNA ligand recognition receptor. In consequence, interference with NOD1-mediated signaling significantly weakens the inflammatory response to dsRNA. These results add a new level of complexity to the understanding of the cross talk between different classes of pattern recognition receptors and may be related to certain complications of chronic hepatitis C virus infection.
Collapse
Affiliation(s)
- Serena Vegna
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Damien Gregoire
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Marie Moreau
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Patrice Lassus
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - David Durantel
- INSERM, U1052, Cancer Research Center of Lyon, University of Lyon, Lyon, France
| | - Eric Assenat
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
- Service d'Oncologie Médicale, CHU St. Eloi, Montpellier, France
| | - Urszula Hibner
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Yannick Simonin
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Université de Montpellier, Montpellier, France
| |
Collapse
|
8
|
Durmuş S, Çakır T, Özgür A, Guthke R. A review on computational systems biology of pathogen-host interactions. Front Microbiol 2015; 6:235. [PMID: 25914674 PMCID: PMC4391036 DOI: 10.3389/fmicb.2015.00235] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/10/2015] [Indexed: 12/27/2022] Open
Abstract
Pathogens manipulate the cellular mechanisms of host organisms via pathogen-host interactions (PHIs) in order to take advantage of the capabilities of host cells, leading to infections. The crucial role of these interspecies molecular interactions in initiating and sustaining infections necessitates a thorough understanding of the corresponding mechanisms. Unlike the traditional approach of considering the host or pathogen separately, a systems-level approach, considering the PHI system as a whole is indispensable to elucidate the mechanisms of infection. Following the technological advances in the post-genomic era, PHI data have been produced in large-scale within the last decade. Systems biology-based methods for the inference and analysis of PHI regulatory, metabolic, and protein-protein networks to shed light on infection mechanisms are gaining increasing demand thanks to the availability of omics data. The knowledge derived from the PHIs may largely contribute to the identification of new and more efficient therapeutics to prevent or cure infections. There are recent efforts for the detailed documentation of these experimentally verified PHI data through Web-based databases. Despite these advances in data archiving, there are still large amounts of PHI data in the biomedical literature yet to be discovered, and novel text mining methods are in development to unearth such hidden data. Here, we review a collection of recent studies on computational systems biology of PHIs with a special focus on the methods for the inference and analysis of PHI networks, covering also the Web-based databases and text-mining efforts to unravel the data hidden in the literature.
Collapse
Affiliation(s)
- Saliha Durmuş
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University, KocaeliTurkey
| | - Tunahan Çakır
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University, KocaeliTurkey
| | - Arzucan Özgür
- Department of Computer Engineering, Boǧaziçi University, IstanbulTurkey
| | - Reinhard Guthke
- Leibniz Institute for Natural Product Research and Infection Biology – Hans-Knoell-Institute, JenaGermany
| |
Collapse
|
9
|
Li Z, Linghu E, Cheng J. Screening of hepatocyte proteins binding with the middle surface protein of the hepatitis B virus by the yeast two-hybrid system. Mol Med Rep 2014; 9:2342-6. [PMID: 24676405 DOI: 10.3892/mmr.2014.2069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 02/20/2014] [Indexed: 11/05/2022] Open
Abstract
The effect of the middle hepatitis B virus surface protein (MHBs) remains to be elucidated. To investigate the biological function of the MHBs protein, the present study performed yeast two-hybrid screening to search for proteins that interact with the MHBs protein in hepatocytes. The bait plasmid expressing the MHBs protein was constructed by cloning the gene of the MHBs protein into pGBKT7, then the recombinant plasmid DNA was transformed into AH109 yeast (a type). The transformed yeast AH109 was mated with yeast Y187 (α type) containing the liver cDNA library plasmid in 2X yeast peptone dextrose adenine (YPDA) medium. The mated diploid yeast was plated on quadruple dropout medium (SD/-Trp-Leu-His-Ade) containing X-α-gal for selection and screening. Following extracting and sequencing of the plasmids from positive (blue) colonies, the sequence analysis was conducted and analyzed by bioinformatics methods. Two colonies were selected and sequenced. Among them, one was the human DNA sequence from the clone RP11-490D19 on chromosome 9 and the other was homo sapiens 12 BAC RP11-180M15 (Roswell Park Cancer Institute Human BAC Library). The yeast two-hybrid system is an effective method for identifying hepatocyte proteins that interact with MHBs. The MHBs protein binds with different proteins suggesting that it has multiple functions in vivo.
Collapse
Affiliation(s)
- Zhiqun Li
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Enqiang Linghu
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jun Cheng
- Institute of Infectious Diseases, Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| |
Collapse
|
10
|
Kasama Y, Mizukami T, Kusunoki H, Peveling-Oberhag J, Nishito Y, Ozawa M, Kohara M, Mizuochi T, Tsukiyama-Kohara K. B-cell-intrinsic hepatitis C virus expression leads to B-cell-lymphomagenesis and induction of NF-κB signalling. PLoS One 2014; 9:e91373. [PMID: 24651473 PMCID: PMC3961254 DOI: 10.1371/journal.pone.0091373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/10/2014] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) infection leads to the development of hepatic diseases, as well as extrahepatic disorders such as B-cell non-Hodgkin's lymphoma (B-NHL). To reveal the molecular signalling pathways responsible for HCV-associated B-NHL development, we utilised transgenic (Tg) mice that express the full-length HCV genome specifically in B cells and develop non-Hodgkin type B-cell lymphomas (BCLs). The gene expression profiles in B cells from BCL-developing HCV-Tg mice, from BCL-non-developing HCV-Tg mice, and from BCL-non-developing HCV-negative mice were analysed by genome-wide microarray. In BCLs from HCV-Tg mice, the expression of various genes was modified, and for some genes, expression was influenced by the gender of the animals. Markedly modified genes such as Fos, C3, LTβR, A20, NF-κB and miR-26b in BCLs were further characterised using specific assays. We propose that activation of both canonical and alternative NF-κB signalling pathways and down-regulation of miR-26b contribute to the development of HCV-associated B-NHL.
Collapse
Affiliation(s)
- Yuri Kasama
- Department of Experimental Phylaxiology, Faculty of Life Sciences, Kumamoto University, Kumamoto-shi, Kumamoto, Japan
| | - Takuo Mizukami
- Department of Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi-Murayama-shi, Tokyo, Japan
| | - Hideki Kusunoki
- Department of Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi-Murayama-shi, Tokyo, Japan
| | | | - Yasumasa Nishito
- Center for Microarray Analysis, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Tokyo, Japan
| | - Makoto Ozawa
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
- Laboratory of Animal Hygiene, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Tokyo, Japan
| | - Toshiaki Mizuochi
- Department of Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi-Murayama-shi, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
- Laboratory of Animal Hygiene, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
- * E-mail:
| |
Collapse
|
11
|
Machida K. Tumor-initiating stem-like cells and drug resistance: carcinogenesis through Toll-like receptors, environmental factors, and virus. Drug Deliv Transl Res 2013; 3:152-64. [PMID: 25787983 PMCID: PMC10578060 DOI: 10.1007/s13346-012-0115-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neoplasms contain distinct subpopulations of cells known as tumor-initiating stem-like cells (TICs) that have been identified as key drivers of tumor growth and malignant progression with drug resistance. Stem cells normally proliferate through self-renewing divisions in which the two daughter cells differ markedly in their proliferative potential, with one displaying the differentiation phenotypes and another retaining self-renewing activity. Therefore, understanding the molecular mechanisms of hepatocarcinogenesis will be required for the eventual development of improved therapeutic modalities for treating hepatocellular carcinoma (HCC). Hepatitis C virus (HCV) and hepatitis B virus is a major cause of HCC. Compelling epidemiologic evidence identifies obesity and alcohol as co-morbidity factors that can increase the risk of HCV patients for HCC, especially in alcoholics or obese patients. The mechanisms underlying liver oncogenesis, and how environmental factors contribute to this process, are not yet understood. The HCV-Toll-like receptor 4 (TLR4)-Nanog signaling network is established since alcohol/obesity-associated endotoxemia then activates TLR4 signaling, resulting in the induction of the stem cell marker Nanog expression and liver tumors. Liver TICs are highly sensitized to leptin and exposure of TICs to leptin increases the expression and activity of an intrinsic pluripotency-associated transcriptional network comprised of signal transducer and activator of transcription 3, SOX2, OCT4, and Nanog. Stimulation of the pluripotency network may have significant implications for hepatocellular oncogenesis via genesis and maintenance of TICs. It is important to understand how HCV induces liver cancer through genesis of TICs so that better prevention and treatment can be found. This article reviews the oncogenic pathways to generate TICs.
Collapse
Affiliation(s)
- Keigo Machida
- Department of Molecular Microbiology and Immunology, Research Center for ALPD and Cirrhosis, University of Southern California School of Medicine, 503C-HMR, Los Angeles, CA, 90033, USA,
| |
Collapse
|
12
|
Liu Y, Yin Q, Yuan Y, Yang W, Jiang C, Huang C. Infectomics Screening for Novel Antiviral Drug Targets. Drug Dev Res 2012. [PMCID: PMC7163650 DOI: 10.1002/ddr.21027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Copyright 2012 Wiley-Liss, Inc., A Wiley Company Infectomics, a novel way to globally and comprehensively understand the interactions between microbial pathogens and their hosts, has significantly expanded understanding of the microbial infections. The infectomics view of viral–host interactions on the viral perspective principally focuses on gene acquisition, deletion, and point mutation, while traditional antiviral drug discovery concentrates on viral encoding proteins. Recently, high‐throughput technologies, such as mass spectrometry‐based proteomics, activity‐based protein profiling, microarray analysis, yeast two‐hybrid assay, small interfering RNA screening, and micro RNA profiling, have been gradually employed in the research of virus–host interactions. Besides, signaling pathways and cellular processes involved in viral–host interactions provide new insights of infectomics in antiviral drug discovery. In this review, we summarize related infectomics approaches in the studies of virus–host interactions, which shed light on the development of novel antiviral drug targets screening.
Collapse
Affiliation(s)
- Yuan Liu
- The State Key Laboratory of Biotherapy; West China Hospital, West China, Sichuan University; Chengdu; 610041; China
| | - Qi Yin
- The State Key Laboratory of Biotherapy; West China Hospital, West China, Sichuan University; Chengdu; 610041; China
| | - Yao Yuan
- The State Key Laboratory of Biotherapy; West China Hospital, West China, Sichuan University; Chengdu; 610041; China
| | - Wenyong Yang
- The State Key Laboratory of Biotherapy; West China Hospital, West China, Sichuan University; Chengdu; 610041; China
| | - Chuangui Jiang
- The State Key Laboratory of Biotherapy; West China Hospital, West China, Sichuan University; Chengdu; 610041; China
| | - Canhua Huang
- The State Key Laboratory of Biotherapy; West China Hospital, West China, Sichuan University; Chengdu; 610041; China
| |
Collapse
|
13
|
Akcam FZ, Tigli A, Kaya O, Ciris M, Vural H. Cytokine levels and histopathology in chronic hepatitis B and chronic hepatitis C. J Interferon Cytokine Res 2012; 32:570-4. [PMID: 23067363 DOI: 10.1089/jir.2012.0048] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The changes in balance of cytokine profile may result in either recovery or persistence of hepatitis B virus (HBV) and hepatitis C virus (HCV) infections. This study aims to reveal a possible correlation between cytokine levels, ie, tumor necrosis factor (TNF)-α; interferon-gamma (IFN-γ); interleukin (IL)-10, IL-18, and transforming growth factor-beta (TGF-β); and Ishak score or fibrosis in patients with chronic hepatitis B (CHB) or chronic hepatitis C (CHC). Fifty patients with CHB (n=25), CHC (n=25), and the control group of subjects with negative hepatitis B and C serology (n=30) were included in the study. Patients who did not agree to participate in the study were excluded. Serum cytokine levels were measured by ELISA. Liver biopsies from the patients were also taken for pathological analyses by the same pathologist. The serum levels of TNF-α, IL-10, and IL-18 in the hepatitis C group were significantly high compared with those of the control group (P=0.017, P=0.001, and P=0.004 respectively), but, only IL-10 levels in the hepatitis B group were significantly high (P=0.001). These groups did not show any significant difference with respect to IFN-γ or TGF-β levels. In patients with CHB or CHC, there was a significant correlation (P=0.000) between TNF-α and Ishak score or fibrosis; but no such correlation was found with IFN-γ, IL-10, IL-18, or TGF-β. Result of the current study indicated that cytokine activities were important indicators of clinical severity and progression of HBV- and HCV infections. Further investigations on possible effects of cytokines on hepatocellular damage and fibrosis should be done to arrange new immunopathological approaches to viral hepatitis.
Collapse
Affiliation(s)
- Fusun Zeynep Akcam
- Department of Infectious Diseases and Clinical Microbiology, Suleyman Demirel University, Isparta 32040, Turkey.
| | | | | | | | | |
Collapse
|
14
|
Yu JZ, Wu XL, Yu B, Dai CQ, Liu KL, Guo-Qiang Q, Zhou GX, Lu SH, Ju DH, Chen XY. The suppression effects of desacetyluvaricin on hepatocellular carcinoma and its possible mechanism. Pharmacogn Mag 2012; 8:225-30. [PMID: 23060697 PMCID: PMC3466458 DOI: 10.4103/0973-1296.99288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/12/2011] [Accepted: 08/02/2012] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To investigate the anticancer effects of desacetyluvaricin (DES) on hepatocellular carcinoma (HCC) in vitro, and to study its mechanism. MATERIALS AND METHODS Using DES and cisplatin (DDP) to intervene the cell lines of hepatocarcinoma G2.2.15 (HepG2.2.15) and HepG2, by detecting the expression of HBxAg by immunofluorescence method, the cell cycle and apoptosis by flow cytometry method (FCM), and expression of NF-κB protein by ELISA. RESULTS DES and DDP showed to suppress proliferation of HepG2.2.15 and HepG2; they increase the S-phase cells and decrease G2/M phase cells. DES and DDP both could promote the apoptosis and reduce the expression of NF-κB on the cell line. DES and DDP both can suppress the expression of HbxAg in HepG2.2.15. There were no statistical differences of the above results between these two drugs (P > 0.05). CONCLUSIONS DES possesses anticancer effect on hepatocarcinoma. The possible mechanism might be due to promotion the apoptosis of the cancer cells, and downregulate the expression of HBx andNF-κB protein. DES is a kind of natural products, Because of the lighter clinical side effects; our observations suggest that DES has the potential to be explored as an effective anticancer agent for HCC.
Collapse
Affiliation(s)
- Jian Zhong Yu
- Department of Internal Medicine, The Hospital of Beijing Tong Ren Tang, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kwofie SK, Schaefer U, Sundararajan VS, Bajic VB, Christoffels A. HCVpro: Hepatitis C virus protein interaction database. INFECTION GENETICS AND EVOLUTION 2011; 11:1971-7. [PMID: 21930248 DOI: 10.1016/j.meegid.2011.09.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 08/24/2011] [Accepted: 09/02/2011] [Indexed: 02/07/2023]
|
16
|
Biology and signal transduction pathways of the Lymphotoxin-αβ/LTβR system. Cytokine Growth Factor Rev 2011; 22:301-10. [DOI: 10.1016/j.cytogfr.2011.11.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
Abstract
Chronic liver damage caused by viral infection, alcohol, or obesity can result in increased risk for hepatocellular carcinoma (HCC). Ample epidemiological evidence suggests that there is a strong synergism between hepatitis C virus (HCV) and alcoholic liver diseases (ALD). The Toll-like receptor (TLR) signaling pathway is upregulated in chronic liver diseases. Alcoholism is associated with endotoxemia that stimulates expression of proinflammatory cytokine expression and inflammation in the liver and fat tissues. Recent studies of HCC have centered on cancer-initiating stem cell (CSC), including detection of CSC in cancer, identification of CSC markers, and isolation of CSC from human HCC cell lines. Synergism between alcohol and HCV may lead to liver tumorigenesis through TLR signaling.
Collapse
|
18
|
Lymphotoxin's link to carcinogenesis: friend or foe? from lymphoid neogenesis to hepatocellular carcinoma and prostate cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:231-49. [PMID: 21153327 DOI: 10.1007/978-1-4419-6612-4_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Pezacki JP, Singaravelu R, Lyn RK. Host-virus interactions during hepatitis C virus infection: a complex and dynamic molecular biosystem. MOLECULAR BIOSYSTEMS 2010; 6:1131-42. [PMID: 20549003 DOI: 10.1039/b924668c] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The hepatitis C virus (HCV) is a global health issue with no vaccine available and limited clinical treatment options. Like other obligate parasites, HCV requires host cellular components of an infected individual to propagate. These host-virus interactions during HCV infection are complex and dynamic and involve the hijacking of host cell environments, enzymes and pathways. Understanding this unique molecular biosystem has the potential to yield new and exciting strategies for therapeutic intervention. Advances in genomics and proteomics have opened up new possibilities for the rapid measurement of global changes at the transcriptional and translational levels during infection. However, these techniques only yield snapshots of host-virus interactions during HCV infection. Other new methods that involve the imaging of biomolecular interactions during HCV infection are required to identify key interactions that may be transient and dynamic. Herein we highlight systems biology based strategies that have helped to identify key host-virus interactions during HCV replication and infection. Novel biophysical tools are also highlighted for identification and visualization of activities and interactions between HCV and its host hepatocyte. As some of these methods mature, we expect them to pave the way forward for further exploration of this complex biosystem and elucidation of mechanisms for HCV pathogenesis and carcinogenesis.
Collapse
Affiliation(s)
- John Paul Pezacki
- Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Dr., Ottawa, Ontario, Canada.
| | | | | |
Collapse
|
20
|
Tumor necrosis factor receptor 1 expression is upregulated in dendritic cells in patients with chronic HCV who respond to therapy. HEPATITIS RESEARCH AND TREATMENT 2010; 2010:429243. [PMID: 21152182 PMCID: PMC2989713 DOI: 10.1155/2010/429243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/28/2010] [Accepted: 05/31/2010] [Indexed: 11/18/2022]
Abstract
The present studies assessed the level of tumor necrosis factor receptor (TNFR) expression in peripheral blood mononuclear cells (PBMCs) subsets from patients with chronic HCV undergoing interferon α/ribavirin-based therapy (Ifn/R). Methods. TNFR family member mRNA expression was determined using quantitative real-time PCR assays (RTPCRs) in PBMC from 39 HCV+ patients and 21 control HCV− patients. Further subset analysis of HCV + patients (untreated (U), sustained virological responders (SVR), and nonresponders (NR)/relapsers (Rel)) PBMC was performed via staining with anti-CD123, anti-CD33, anti-TNFR1 or via RTPCR for TNFR1 mRNA. Results. A similar level of TNFR1 mRNA in PBMC from untreated HCV+ genotype 1 patients and controls was noted. TNFR1 and TNFR2 mRNA levels in PBMC from HCV+ patients with SVR were statistically different than levels in HCV(−) patients. A significant difference was noted between the peak values of TNFR1 of the CD123+ PBMC isolated from SVR and the NR/Rel. Conclusion. Upregulation of TNFR1 expression, occurring in a specific subset of CD123+ dendritic cells, appeared in HCV+ patients with SVR.
Collapse
|
21
|
Strosberg AD, Kota S, Takahashi V, Snyder JK, Mousseau G. Core as a novel viral target for hepatitis C drugs. Viruses 2010; 2:1734-1751. [PMID: 21994704 PMCID: PMC3185734 DOI: 10.3390/v2081734] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 08/06/2010] [Accepted: 08/16/2010] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) infects over 130 million people worldwide and is a major cause of liver disease. No vaccine is available. Novel specific drugs for HCV are urgently required, since the standard-of-care treatment of pegylated interferon combined with ribavirin is poorly tolerated and cures less than half of the treated patients. Promising, effective direct-acting drugs currently in the clinic have been described for three of the ten potential HCV target proteins: NS3/NS4A protease, NS5B polymerase and NS5A, a regulatory phosphoprotein. We here present core, the viral capsid protein, as another attractive, non-enzymatic target, against which a new class of anti-HCV drugs can be raised. Core plays a major role in the virion's formation, and interacts with several cellular proteins, some of which are involved in host defense mechanisms against the virus. This most conserved of all HCV proteins requires oligomerization to function as the organizer of viral particle assembly. Using core dimerization as the basis of transfer-of-energy screening assays, peptides and small molecules were identified which not only inhibit core-core interaction, but also block viral production in cell culture. Initial chemical optimization resulted in compounds active in single digit micromolar concentrations. Core inhibitors could be used in combination with other HCV drugs in order to provide novel treatments of Hepatitis C.
Collapse
Affiliation(s)
- Arthur Donny Strosberg
- Department of Infectology, The Scripps Research Institute-Scripps Florida, 130 Scripps Way, Jupiter, FL-33458, USA; E-Mails: (S.K.); (V.T.); (G.M.)
| | - Smitha Kota
- Department of Infectology, The Scripps Research Institute-Scripps Florida, 130 Scripps Way, Jupiter, FL-33458, USA; E-Mails: (S.K.); (V.T.); (G.M.)
| | - Virginia Takahashi
- Department of Infectology, The Scripps Research Institute-Scripps Florida, 130 Scripps Way, Jupiter, FL-33458, USA; E-Mails: (S.K.); (V.T.); (G.M.)
| | - John K. Snyder
- Department of Chemistry, The Center for Chemical Methodology and Library Development, Boston University, Boston, MA 02215, USA; E-Mail:
| | - Guillaume Mousseau
- Department of Infectology, The Scripps Research Institute-Scripps Florida, 130 Scripps Way, Jupiter, FL-33458, USA; E-Mails: (S.K.); (V.T.); (G.M.)
| |
Collapse
|
22
|
Sanjo H, Zajonc DM, Braden R, Norris PS, Ware CF. Allosteric regulation of the ubiquitin:NIK and ubiquitin:TRAF3 E3 ligases by the lymphotoxin-beta receptor. J Biol Chem 2010; 285:17148-55. [PMID: 20348096 PMCID: PMC2878066 DOI: 10.1074/jbc.m110.105874] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 03/01/2010] [Indexed: 12/28/2022] Open
Abstract
The lymphotoxin-beta receptor (LTbetaR) activates the NF-kappaB2 transcription factors, p100 and RelB, by regulating the NF-kappaB-inducing kinase (NIK). Constitutive proteosomal degradation of NIK limits NF-kappaB activation in unstimulated cells by the ubiquitin:NIK E3 ligase comprised of subunits TNFR-associated factors (TRAF)3, TRAF2, and cellular inhibitor of apoptosis (cIAP). However, the mechanism releasing NIK from constitutive degradation remains unclear. We found that insertion of a charge-repulsion mutation in the receptor-binding crevice of TRAF3 ablated binding of both LTbetaR and NIK suggesting a common recognition site. A homologous mutation in TRAF2 inhibited cIAP interaction and blocked NIK degradation. Furthermore, the recruitment of TRAF3 and TRAF2 to the ligated LTbetaR competitively displaced NIK from TRAF3. Ligated LTbetaR complexed with TRAF3 and TRAF2 redirected the specificity of the ubiquitin ligase reaction to polyubiquitinate TRAF3 and TRAF2, leading to their proteosomal degradation. Stimulus-dependent degradation of TRAF3 required the RING domain of TRAF2, but not of TRAF3, implicating TRAF2 as a key E3 ligase in TRAF turnover. The combined action of competitive displacement of NIK and TRAF degradation halted NIK turnover, and promoted its association with IKKalpha and signal transmission. These results indicate the LTbetaR modifies the ubiquitin:NIK E3 ligase, and also acts as an allosteric regulator of the ubiquitin:TRAF E3 ligase.
Collapse
Affiliation(s)
| | - Dirk M. Zajonc
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037
| | | | | | | |
Collapse
|
23
|
Yan XB, Chen Z, Brechot C. Associations among Genotype 1b Hepatitis C Virus Core Protein, Protein Kinase R, and Signal Transducer and Activator of Transcription 3. HEPATITIS MONTHLY 2010; 10:275-84. [PMID: 22312393 PMCID: PMC3271320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 07/22/2010] [Accepted: 09/17/2010] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS Because hepatitis C virus (HCV) core protein (Core), protein kinase R (PKR), and signal transducer and activator of transcription 3 (STAT3) all play relevant roles in the pathogenesis of HCV, persistent infection and hepatocellular carcinoma (HCC) and PKR may interact with HCV Core. In this study, we further investigate the associations among HCV Core, PKR, and STAT3 and the mechanisms involved in these interactions. MATERIALS AND METHODS Expression levels of HCV Core, PKR, eukaryotic initiation factor 2 (eIF-2α), phosphorylated eIF- 2α (p-eIF-2α), STAT3, and phosphorylated-STAT3 (p-STAT3) were compared between Huh-7 and replicon cell-Huh-7 cells harboring the full length of genotype 1b HCV genomes. Co-immunoprecipitation and glutathione S-transferase (GST) pull-down assay were conducted for HCV Core, PKR, and STAT3. RESULTS HCV may have induced the expression of STAT3 and the activity of PKR (p-eIF-2α). HCV Core, STAT3, and PKR appear to have interacted with one another. The N-terminal 1-126 amino acid (aa) of HCV Core contributed to an interaction between HCV Core and STAT3, and only full-length PKR bound to STAT3 and p-STAT3. CONCLUSIONS These findings suggest that HCV Core, PKR, and STAT3 can interact with each other. Specifically, HCV Core may play its role through both PKR and STAT3. Alternatively, HCV Core's binding to and activation of STAT3 might be due to the interaction between HCV Core and PKR. The distinct interactions among these three molecules are important and may reveal a new molecular mechanism in the pathogenesis of HCV-persistent infection and HCV-related HCC.
Collapse
Affiliation(s)
- Xue bing Yan
- Department of Infectious Diseases, The First Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu Province, China,Corresponding author at: Dr. Xue-bing Yan, Department of Infectious Diseases, The First Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu Province, China. Tel.: +51-685802180, Fax: +51-685802180, E-mail:
| | - Zhi Chen
- Institute of Infectious Diseases, the First Affiliated Hospital, College of Medical Science, Key Laboratory of Ministry Health, Zhejian
| | | |
Collapse
|
24
|
Hu Z, Muroyama R, Kowatari N, Chang J, Omata M, Kato N. Characteristic mutations in hepatitis C virus core gene related to the occurrence of hepatocellular carcinoma. Cancer Sci 2009; 100:2465-8. [PMID: 20175782 PMCID: PMC11158212 DOI: 10.1111/j.1349-7006.2009.01338.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 08/13/2009] [Accepted: 08/20/2009] [Indexed: 12/21/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection often results in hepatocellular carcinoma (HCC). Previous studies have shown that there might be some characteristic mutations in the core region of HCV related to HCC. Thus, we downloaded and analyzed HCV genotype 1b core gene sequences from HCV databases online to identify them. Based on the information of the sequences, 63 from patients with HCC and 188 from non-HCC were enrolled into our analysis. Then, the nucleotides at each position were compared by chi(2)-test between the two groups, and 24 polymorphisms were found to be associated with HCC. Further analysis of these 24 polymorphisms by logistic regression indicated that eight were significantly related to the increased HCC risk: A028C, G209A, C219U/A, U264C, A271C/U, C378U/A, G435A/C, and G481A. Moreover, U303C/A was associated with the decreased HCC risk. These mutations could bring about four amino acid substitutions: K10Q, R70Q, M91L, and G161S. In conclusion, eight characteristic mutations in the HCV-1b core gene related to the occurrence of HCC were identified. The structural and functional alterations of core protein due to these mutations and the relationship with the occurrence of HCC need to be further studied.
Collapse
Affiliation(s)
- Zhongjie Hu
- Unit of Disease Control Genome Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Ferrari C, Mondelli M. Immune Mechanisms of Viral Clearance and Disease Pathogenesis During Viral Hepatitis. THE LIVER 2009:835-857. [DOI: 10.1002/9780470747919.ch51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Chen YR, Chen TY, Zhang SL, Lin SM, Zhao YR, Ye F, Zhang X, Shi L, Dang SS, Liu M. Identification of a novel protein binding to hepatitis C virus core protein. J Gastroenterol Hepatol 2009; 24:1300-4. [PMID: 19486448 DOI: 10.1111/j.1440-1746.2009.05846.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND Hepatitis C virus (HCV) core protein is a multi-functional viral protein that interacts with several target proteins of both viral and cellular origin. AIM AND METHODS To gain insight into the mechanism of action of HCV core protein, we used a yeast two-hybrid system to identify the core protein-interacting cellular targets. RESULTS A cDNA clone encoding an aspartoacylase was obtained, termed aspartoacylase 3 (ACY3). Interaction between ACY3 and HCV core protein was verified using a co-immunoprecipitation assay in vitro, and a mammalian two-hybrid system in vivo. Fluorescence microscopy showed green fluorescence protein-fused ACY3 localized in the cytoplasm. CONCLUSION Our data suggest that ACY3 is an HCV core binding protein, which may play a role in the development of HCV-associated diseases.
Collapse
Affiliation(s)
- Yun-Ru Chen
- The First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Liu L, Li Y, Lin J, Liang Q, Sheng X, Wu J, Huang R, Liu S, Li Y. Connexin43 interacts with Caveolin-3 in the heart. Mol Biol Rep 2009; 37:1685-91. [PMID: 19544087 DOI: 10.1007/s11033-009-9584-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 06/08/2009] [Indexed: 02/05/2023]
Abstract
Gap junctions (GJs), collections of multiple intercellular channels between neighboring cells, are specialized channels facilitating intercellular electrical and chemical communication. GJs are important for synchronizing coupling and coordinated contraction in the heart, and are crucial regulators of heart gene transcription, cardiac development, and protection of ischemic cardiomyocytes through second messenger communication. Identification of proteins that interact with Connexin43 (Cx43), the predominant protein in cardiac GJs, may contribute to the understanding of GJ functional regulation. Using a yeast two-hybrid system, we identified Caveolin-3 (Cav3) as a new Cx43-interacting protein. This interaction was confirmed by co-immunoprecipitation and co-localization experiments. CX43 interacts with Cav3, suggesting that Cav3 may participate in the functional regulation of GJs.
Collapse
Affiliation(s)
- Limei Liu
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, 57 Changping Road, 515041, Shantou, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tran G. The role of hepatitis C virus in the pathogenesis of hepatocellular carcinoma. ACTA ACUST UNITED AC 2008. [DOI: 10.1093/biohorizons/hzn020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
29
|
|
30
|
Abstract
In recent years, the effects of hepatitis C virus (HCV) proteins on hepatocarcinogenesis have undergone intense investigations. The potentially oncogenic proteins include at least three HCV proteins: core (C) protein, NS3, and NS5A. Several authors indicated relationships between subcellular localization, concentration, a specific molecular form of the proteins (full length, truncated, phosphorylated), the presence of specific domains (the nuclear localization signal homologous to e.g. Bcl-2) and their effects on the mechanisms linked to oncogenesis. The involvement of all the proteins has been described as being in control of the cell cycle, through interactions with key proteins of the process (p53, p21, cyclins, proliferating cell nuclear antigen), transcription factors, proto-oncogenes, growth factors/cytokines and their receptors, and proteins linked to the apoptotic process. Untilnow, the involvement of the core protein of HCV in liver carcinogenesis is the most recognized. One of the most common proteins affected by HCV proteins is the p53 tumor-suppressor protein. The p21/WAF1 gene is a major target of p53, and the effect of HCV proteins on the gene is frequently considered in parallel. The results of studies on the effects of HCV proteins on the apoptotic process are controversial. This work summarizes the information collected thus far in the field of HCV molecular virology and principal intracellular signaling pathways in which HCV oncogenic proteins are involved.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, Medical University, Poznań, Poland
| | | |
Collapse
|
31
|
Krekulová L, Rehák V, Riley LW. Structure and functions of hepatitis C virus proteins: 15 years after. Folia Microbiol (Praha) 2007; 51:665-80. [PMID: 17455808 DOI: 10.1007/bf02931636] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since its discovery in 1988, the hepatitis C virus (HCV) has become a hot topic of research by many groups around the world. This globally spread infectious agent is responsible for a large proportion of chronic viral hepatitides. The clue to halting the hepatitis C pandemic may be the detailed understanding of the virus structure, its replication mechanism, and the exact functions of the various proteins. Such understanding could enable the development of new antivirals targeted against hepatitis C virus and possibly an effective vaccine. This review recaps the current knowledge about the HCV genome 15 years after its discovery. The structure and function of particular viral structural (core, E1, E2) and nonstructural (NS2, NS3, NS4, NS5) proteins and noncoding regions known to date are described. With respect to frequent conflicting reports from different research groups, results reproducibly demonstrated by independent investigators are emphasized. Owing to many obstacles and limitations inherent in doing research on this noteworthy virus, the current knowledge is incomplete and the answers to many important questions are to be expected in the future.
Collapse
Affiliation(s)
- L Krekulová
- Hepatology, Nusle Clinic, Remedis--Nusle Clinic, Prague, Czechia
| | | | | |
Collapse
|
32
|
Abstract
Apoptosis is central for the control and elimination of viral infections. In chronic hepatitis C virus (HCV) infection, enhanced hepatocyte apoptosis and upregulation of the death inducing ligands CD95/Fas, TRAIL and TNFα occur. Nevertheless, HCV infection persists in the majority of patients. The impact of apoptosis in chronic HCV infection is not well understood. It may be harmful by triggering liver fibrosis, or essential in interferon (IFN) induced HCV elimination. For virtually all HCV proteins, pro- and anti-apoptotic effects have been described, especially for the core and NS5A protein. To date, it is not known which HCV protein affects apoptosis in vivo and whether the infectious virions act pro- or anti-apoptotic. With the availability of an infectious tissue culture system, we now can address pathophysiologically relevant issues. This review focuses on the effect of HCV infection and different HCV proteins on apoptosis and of the corresponding signaling cascades.
Collapse
Affiliation(s)
- Richard Fischer
- Department of Internal Medicine II, University of Freiburg, Hugstetter Strasse 55, D-79106 Freiburg, Germany.
| | | | | |
Collapse
|
33
|
Norris PS, Ware CF. The LT beta R signaling pathway. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 597:160-72. [PMID: 17633025 DOI: 10.1007/978-0-387-70630-6_13] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The lymphotoxin-beta receptor (LTbetaR, TNFRSF3) signaling pathway activates gene transcription programs and cell death important in immune development and host defense. The TNF receptor associated factors (TRAF)-2, 3 and 5 function as adaptors linking LTbetaR signaling targets. Interestingly, TRAF deficient mice do not phenocopy mice deficient in components of the LTbetaR pathway, presenting a conundrum. Here, an update of our understanding and models of the LTbetaR signaling pathway are reviewed, with a focus on this conundrum.
Collapse
Affiliation(s)
- Paula S Norris
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, San Diego, California 92121, USA
| | | |
Collapse
|
34
|
Ng TI, Mo H, Pilot-Matias T, He Y, Koev G, Krishnan P, Mondal R, Pithawalla R, He W, Dekhtyar T, Packer J, Schurdak M, Molla A. Identification of host genes involved in hepatitis C virus replication by small interfering RNA technology. Hepatology 2007; 45:1413-21. [PMID: 17518369 DOI: 10.1002/hep.21608] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
UNLABELLED Hepatitis C virus (HCV) replication is highly dependent on host cell factors. Identification of these host factors not only facilitates understanding of the biology of HCV infection but also enables the discovery of novel targets for anti-HCV therapy. To identify host genes important for HCV RNA replication, we screened a library of small interfering RNA (siRNA) that targets approximately 4,000 human genes in Huh7-derived EN5-3 cells harboring an HCV subgenomic replicon with the nonstructural region NS3-NS5B from the 1b-N strain. Nine cellular genes that potentially regulate HCV replication were identified in this screen. Silencing of these genes resulted in inhibition of HCV replication by more than 60% and exhibited minimal toxicity. Knockdown of host gene expression by these siRNAs was confirmed at the RNA level and, in some instances, at the protein level. The level of siRNA silencing of these host genes correlated well with inhibition of HCV. These genes included those that encoded a G-protein coupled receptor (TBXA2R), a membrane protein (LTbeta), an adapter protein (TRAF2), 2 transcription factors (RelA and NFkappaB2), 2 protein kinases (MKK7 and SNARK), and 2 closely related transporter proteins (SLC12A4 and SLC12A5). Of interest, some of these genes are members of the tumor necrosis factor/lymphotoxin signaling pathway. CONCLUSION Findings of this study may provide important information for understanding HCV replication. In addition, these cellular genes may constitute a novel set of targets for HCV antiviral therapy.
Collapse
Affiliation(s)
- Teresa I Ng
- Global Pharmaceutical Research and Development, Antiviral Research, Abbott Laboratories, 200 Abbott Park Road, Abbott Park, IL 60064, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yan XB, Battaglia S, Boucreux D, Chen Z, Brechot C, Pavio N. Mapping of the interacting domains of hepatitis C virus core protein and the double-stranded RNA-activated protein kinase PKR. Virus Res 2007; 125:79-87. [PMID: 17267064 DOI: 10.1016/j.virusres.2006.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 12/12/2006] [Accepted: 12/14/2006] [Indexed: 01/29/2023]
Abstract
Hepatitis C virus (HCV) core protein has been shown to exhibit several biological properties which suggest an important role in liver pathogenesis and carcinogenesis. During a previous study, we showed that core mutants, isolated from tumour, could directly interact with PKR and maintain it in an activated form. In the present report, we have further investigated this interaction and mapped the core and PKR domains involved. Using glutathion S-transferase fusion protein harbouring the different domains of core or PKR, we determined that the N-terminal 1-58 amino acid (aa) of core protein and the N-terminal 1-180 aa of PKR are responsible for this direct interaction. Using this system we also confirmed that the core-PKR interaction induced PKR autophosphorylation. Furthermore, we found that core protein co-localized and co-immunoprecipitated with PKR in cells expressing a full-length HCV replicon, thus confirming that this interaction occurs when all HCV proteins are expressed. Considering that the activation of PKR has been observed in some cancer cell lines and tissues, it suggests that, depending on the cellular context, PKR may stimulate or inhibit cell proliferation. The precise mapping of core-PKR interaction provides new data to study the molecular mechanism underlying HCV pathogenesis.
Collapse
|
36
|
Abstract
Viral hepatitis is a diffuse inflammatory reaction of the liver caused by hepatotropic viruses. Among the hepatitis viruses, only hepatitis B virus and hepatitis C virus are able to persist in the host and cause chronic hepatitis. In the course of persistent infection, inflammation forms the pathogenetic basis of chronic hepatitis that can lead to nodular fibrosis, which can progress to cirrhosis and, eventually, hepatocellular carcinoma (HCC). Of the different antiviral defense systems employed by the host, apoptosis significantly contributes to the prevention of viral replication, dissemination, and persistence. Pathomorphologic studies have shown acidophilic bodies and hepatocyte dropout, features that are compatible with apoptosis. The number of hepatocytes showing features of apoptosis in patients with chronic hepatitis B and C was found to be higher than in healthy subjects, indicating that apoptosis is involved in the pathogenesis of these diseases. There are various data suggesting that hepatitis B and C viral proteins may modulate apoptosis. Vice versa, mechanisms of apoptosis inhibition might represent central survival strategies employed by the virus which, in the end, may contribute to HCC development. While the expression and retention of viral proteins in hepatocytes may influence the severity and progression of liver disease, the mechanisms of liver injury in viral hepatitis are defined to be due not only to the direct cytopathic effects of viruses, but also to the host immune response to viral proteins expressed by infected hepatocytes. However, the exact role of these observations in relation to pathogenesis remains to be established. The mechanism and systems are complex. This report aims to provide an overview and intends to cite only a small number of pertinent references.
Collapse
Affiliation(s)
- K Herzer
- Deptartment I of Internal Medicine, Mainz University, Mainz, Germany.
| | | | | |
Collapse
|
37
|
Abstract
HCV (hepatitis C virus) has a high propensity to persist and to cause chronic hepatitis C, eventually leading to cirrhosis. Since HCV itself is not cytopathic, liver damage in chronic hepatitis C is commonly attributed to immune-mediated mechanisms. HCV proteins interact with several pathways in the host's immune response and disrupt pathogen-associated pattern recognition pathways, interfere with cellular immunoregulation via CD81 binding and subvert the activity of NK (natural killer) cells as well as CD4(+) and CD8(+) T-cells. Finally, HCV-specific T-cells become increasingly unresponsive and apparently disappear, owing to several possible mechanisms, such as escape mutations in critical viral epitopes, lack of sufficient help, clonal anergy or expansion of regulatory T-cells. The role of neutralizing antibodies remains uncertain, although it is still possible that humoral immunity contributes to bystander damage of virally coated cells via antibody-dependent cellular cytotoxicity. Cytotoxic lymphocytes kill HCV-infected cells via the perforin/granzyme pathway, but also release Fas ligand and inflammatory cytokines such as IFNgamma (interferon gamma). Release of soluble effector molecules helps to control HCV infection, but may also destroy uninfected liver cells and can attract further lymphocytes without HCV specificity to invade the liver. Bystander damage of these non-specific inflammatory cells will expand the tissue damage triggered by HCV infection and ultimately activate fibrogenesis. A clear understanding of these processes will eventually help to develop novel treatment strategies for HCV liver disease, independent from direct inhibition of HCV replication.
Collapse
Affiliation(s)
- Ulrich Spengler
- Department of Internal Medicine 1, University of Bonn, Sigmund-Freud-Strasse 25, Bonn, Germany.
| | | |
Collapse
|
38
|
Sato S, Fukasawa M, Yamakawa Y, Natsume T, Suzuki T, Shoji I, Aizaki H, Miyamura T, Nishijima M. Proteomic profiling of lipid droplet proteins in hepatoma cell lines expressing hepatitis C virus core protein. J Biochem 2006; 139:921-30. [PMID: 16751600 DOI: 10.1093/jb/mvj104] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) core protein has been suggested to play crucial roles in the pathogeneses of liver steatosis and hepatocellular carcinomas due to HCV infection. Intracellular HCV core protein is localized mainly in lipid droplets, in which the core protein should exert its significant biological/pathological functions. In this study, we performed comparative proteomic analysis of lipid droplet proteins in core-expressing and non-expressing hepatoma cell lines. We identified 38 proteins in the lipid droplet fraction of core-expressing (Hep39) cells and 30 proteins in that of non-expressing (Hepswx) cells by 1-D-SDS-PAGE/MALDI-TOF mass spectrometry (MS) or direct nanoflow liquid chromatography-MS/MS. Interestingly, the lipid droplet fraction of Hep39 cells had an apparently lower content of adipose differentiation-related protein and a much higher content of TIP47 than that of Hepswx cells, suggesting the participation of the core protein in lipid droplet biogenesis in HCV-infected cells. Another distinct feature is that proteins involved in RNA metabolism, particularly DEAD box protein 1 and DEAD box protein 3, were detected in the lipid droplet fraction of Hep39 cells. These results suggest that lipid droplets containing HCV core protein may participate in the RNA metabolism of the host and/or HCV, affecting the pathopoiesis and/or virus replication/production in HCV-infected cells.
Collapse
Affiliation(s)
- Shigeko Sato
- Department of Biochemistry and Cell Biology and Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dejardin E. The alternative NF-kappaB pathway from biochemistry to biology: pitfalls and promises for future drug development. Biochem Pharmacol 2006; 72:1161-79. [PMID: 16970925 DOI: 10.1016/j.bcp.2006.08.007] [Citation(s) in RCA: 281] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 08/11/2006] [Accepted: 08/14/2006] [Indexed: 01/08/2023]
Abstract
The past two decades have led to a tremendous work on the transcription factor NF-kappaB and its molecular mechanisms of activation. The nuclear translocation of NF-kappaB is controlled by two main pathways: the classical and the alternative NF-kappaB pathways. The classical NF-kappaB pathway activates the IKK complex that controls the inducible degradation of most IkappaB family members that are IkappaBalpha, IkappaBbeta, IkappaBvarepsilon and p105. The alternative NF-kappaB pathway induces p100 processing and p52 generation through the activation of at least two kinases, which are NIK and IKKalpha. Genetic studies have shown that IKKgamma is dispensable for the alternative pathway, which suggests the existence of an alternative IKKalpha-containing complex. It is noteworthy that activation of particular p52 heterodimers like p52/RelB requires solely the alternative pathway while activation of p52/p65 or p52/c-Rel involves a "hybrid pathway". Among others, LTbetaR, BAFF-R, CD40 and RANK have the ability to induce the alternative pathway. The latter plays some roles in biological functions controlled by these receptors, which are the development of secondary lymphoid organs, the proliferation, survival and maturation of B cell, and the osteoclastogenesis. Exacerbated activation of the alternative pathway is potentially associated to a wide range of disorders like rheumatoid arthritis, ulcerative colitis or B cell lymphomas. Therefore, inhibitors of the alternative pathway could be valuable tools for the treatment of inflammatory disorders and cancers.
Collapse
Affiliation(s)
- Emmanuel Dejardin
- Laboratory of Virology & Immunology, Centre of Biomedical Integrative Genoproteomics (CBIG), University of Liège, Avenue de l'Hôpital, Sart-Tilman, CHU, B23, 4000 Liege, Belgium.
| |
Collapse
|
40
|
Qureshi SA. Hepatitis C virus-biology, host evasion strategies, and promising new therapies on the horizon. Med Res Rev 2006; 27:353-73. [PMID: 16958135 DOI: 10.1002/med.20063] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hepatitis C reduces the quality of life for some 170 million people around the globe and is one of the most prevalent diseases on the planet. It is caused by the hepatitis C virus (HCV) that is replicated by an error-prone polymerase and therefore undergoes rapid evolution. To date, although much has been learned about the biology of HCV, only a partially effective combination therapy comprised of ribavirin and pegylated-interferon-alpha is available to hepatitis C sufferers. Given the prevalence of hepatitis C, together with the fact that almost half the chronically infected HCV patients are refractory to current therapy, there is an urgent need for an efficacious immunoprophylactic that protects individuals from HCV infection, as well as drugs that impede the viral life cycle effectively and eradicate infection. Herein, I provide an overview of the molecular biology of HCV, highlighting the functions of different virally encoded proteins in terms of how they alter signaling pathways of host cell to establish an infection and discuss whether a more promising therapy for treating hepatitis C is anywhere in sight.
Collapse
Affiliation(s)
- Sohail A Qureshi
- Department of Biological & Biomedical Sciences, The Aga Khan University Hospital, Karachi 74800, Pakistan.
| |
Collapse
|
41
|
Abstract
Chronic infection with the hepatitis C virus (HCV) is a major risk factor for the development of hepatocellular carcinoma (HCC) worldwide. The pathogenesis of HCC in HCV infection has extensively been analysed. Hepatitis C virus-induced chronic inflammation and the effects of cytokines in the development of fibrosis and liver cell proliferation are considered as one of the major pathogenic mechanisms. Increasing experimental evidence suggests that HCV contributes to HCC by directly modulating pathways that promote the malignant transformation of hepatocytes. Hepatitis C virus is an RNA virus that does not integrate into the host genome but HCV proteins interact with many host-cell factors well beyond their roles in the viral life cycle and are involved in a wide range of activities, including cell signaling, transcription, cell proliferation, apoptosis, membrane rearrangements, vesicular trafficking and translational regulation. At least four of the HCV gene products, namely HCV core, NS3, NS4B and NS5A, have been shown to exhibit transformation potential in tissue culture and several potentially oncogenic pathways have been shown to be altered by the expression of HCV proteins. Both HCV core and NS5A induce the accumulation of wild-type beta-catenin and the Wnt-beta-catenin pathway emerges as a common target for HCV (and HBV) in human HCCs, also independently from axin/beta-catenin gene mutations. Induction of both endoplasmic reticulum stress and oxidative stress by HCV proteins might also contribute to HCV transformation. Most of the putative transforming functions of the HCV proteins have been defined in artificial cellular systems, which may not be applicable to HCV infection in vivo, and still need to be established in relevant infection and disease models.
Collapse
Affiliation(s)
- M Levrero
- Department of Internal Medicine, University of Rome La Sapienza, Rome, Italy.
| |
Collapse
|
42
|
Saito K, Meyer K, Warner R, Basu A, Ray RB, Ray R. Hepatitis C virus core protein inhibits tumor necrosis factor alpha-mediated apoptosis by a protective effect involving cellular FLICE inhibitory protein. J Virol 2006; 80:4372-9. [PMID: 16611896 PMCID: PMC1472009 DOI: 10.1128/jvi.80.9.4372-4379.2006] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We have previously shown that hepatitis C virus (HCV) core protein modulates multiple cellular processes, including those that inhibit tumor necrosis factor alpha (TNF-alpha)-mediated apoptosis. In this study, we have investigated the signaling mechanism for inhibition of TNF-alpha-mediated apoptosis in human hepatoma (HepG2) cells expressing core protein alone or in context with other HCV proteins. Activation of caspase-3 and the cleavage of DNA repair enzyme poly(ADP-ribose) polymerase were inhibited upon TNF-alpha exposure in HCV core protein-expressing HepG2 cells. In vivo protein-protein interaction studies displayed an association between TNF receptor 1 (TNFR1) and TNFR1-associated death domain protein (TRADD), suggesting that the core protein does not perturb this interaction. A coimmunoprecipitation assay also suggested that HCV core protein does not interfere with the TRADD-Fas-associated death domain protein (FADD)-procaspase-8 interaction. Further studies indicated that HCV core protein expression inhibits caspase-8 activation by sustaining the expression of cellular FLICE (FADD-like interleukin-1beta-converting enzyme)-like inhibitory protein (c-FLIP). Similar observations were also noted upon expression of core protein in context to other HCV proteins expressed from HCV full-length plasmid DNA or a replicon. A decrease in endogenous c-FLIP by specific small interfering RNA induced TNF-alpha-mediated apoptotic cell death and caspase-8 activation. Taken together, our results suggested that the TNF-alpha-induced apoptotic pathway is inhibited by a sustained c-FLIP expression associated with the expression of HCV core protein, which may play a role in HCV-mediated pathogenesis.
Collapse
Affiliation(s)
- Kousuke Saito
- Department of Internal Medicine, Saint Louis University, 3635 Vista Ave., FDT-8N, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Recent studies suggest that liver inflammation in chronic hepatitis C virus (HCV) infection is controlled by several mechanisms, including host regulatory immune responses and viral polypeptides interacting with cells involved in innate and adaptive immunity. This article provides an overview about current thinking on host-pathogen symbiotic relationship in HCV infection and its significance with respect to pathogenesis. Special emphasis is given to regulatory T-cell subsets which have recently received attention and which are thought to play a major role in persistent viral infections such as HCV.
Collapse
Affiliation(s)
- Mario U Mondelli
- Laboratori di Ricerca-Area Infettivologica, IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Italy.
| | | |
Collapse
|
44
|
Abstract
The frequent observation of organized lymphoid structures that resemble secondary lymphoid organs in tissues that are targeted by chronic inflammatory processes, such as autoimmunity and infection, has indicated that lymphoid neogenesis might have a role in maintaining immune responses against persistent antigens. In this Review, we discuss recent progress in several aspects of lymphoid neogenesis, focusing on the similarities with lymphoid tissue development, the mechanisms of induction, functional competence and pathophysiological significance. As more information on these issues becomes available, a better understanding of the role of lymphoid neogenesis in promoting chronic inflammation might eventually lead to new strategies to target immunopathological processes.
Collapse
Affiliation(s)
- Francesca Aloisi
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy.
| | | |
Collapse
|
45
|
Anand S, Wang P, Yoshimura K, Choi IH, Hilliard A, Chen YH, Wang CR, Schulick R, Flies AS, Flies DB, Zhu G, Xu Y, Pardoll DM, Chen L, Tamada K. Essential role of TNF family molecule LIGHT as a cytokine in the pathogenesis of hepatitis. J Clin Invest 2006; 116:1045-51. [PMID: 16557300 PMCID: PMC1409742 DOI: 10.1172/jci27083] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Accepted: 01/31/2006] [Indexed: 12/21/2022] Open
Abstract
LIGHT is an important costimulatory molecule for T cell immunity. Recent studies have further implicated its role in innate immunity and inflammatory diseases, but its cellular and molecular mechanisms remain elusive. We report here that LIGHT is upregulated and functions as a proinflammatory cytokine in 2 independent experimental hepatitis models, induced by concanavalin A and Listeria monocytogenes. Molecular mutagenesis studies suggest that soluble LIGHT protein produced by cleavage from the cell membrane plays an important role in this effect through the interaction with the lymphotoxin-beta receptor (LTbetaR) but not herpes virus entry mediator. NK1.1+ T cells contribute to the production, but not the cleavage or effector functions, of soluble LIGHT. Importantly, treatment with a mAb that specifically interferes with the LIGHT-LTbetaR interaction protects mice from lethal hepatitis. Our studies thus identify a what we believe to be a novel function of soluble LIGHT in vivo and offer a potential target for therapeutic interventions in hepatic inflammatory diseases.
Collapse
MESH Headings
- Animals
- Antigens, Ly
- Antigens, Surface/metabolism
- Concanavalin A/metabolism
- Concanavalin A/pharmacology
- Cytokines/metabolism
- Hepatitis/etiology
- Hepatitis/metabolism
- Inflammation/metabolism
- Lectins, C-Type/metabolism
- Listeria monocytogenes/metabolism
- Listeria monocytogenes/pathogenicity
- Lymphotoxin beta Receptor
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- NK Cell Lectin-Like Receptor Subfamily B
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Member 14
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- Solubility
- Tumor Necrosis Factor Ligand Superfamily Member 14
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Sudarshan Anand
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pu Wang
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kiyoshi Yoshimura
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - In-Hak Choi
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anja Hilliard
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Youhai H. Chen
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chyung-Ru Wang
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard Schulick
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew S. Flies
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dallas B. Flies
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gefeng Zhu
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yanhui Xu
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Drew M. Pardoll
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lieping Chen
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Koji Tamada
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Lin SM, Cheng J, Lu YY, Zhang SL, Yang Q, Chen TY, Liu M, Wang L. Screening and identification of interacting proteins with hepatitis B virus core protein in leukocytes and cloning of new gene C1. World J Gastroenterol 2006; 12:1043-8. [PMID: 16534844 PMCID: PMC4087895 DOI: 10.3748/wjg.v12.i7.1043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2005] [Revised: 07/02/2005] [Accepted: 09/02/2005] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the biological function of HBcAg in pathogenesis of HBV replication in peripheral blood mononuclear cells (PBMCs). METHODS HBcAg region was amplified by polymerase chain reaction (PCR) and HBV HBcAg bait plasmid pGBKT7-HBcAg was constructed by routine molecular biological methods. Then the recombinant plasmid DNA was transformed into yeast AH109. After the HBV core protein was expressed in AH109 yeast strains (Western blot analysis), yeast-two hybrid screening was performed by mating AH109 with Y187 containing leukocyte cDNA library plasmid. Diploid yeast cells were plated on synthetic dropout nutrient medium (SD/-Trp-Leu-His-Ade) (QDO) and synthetic dropout nutrient medium (SD/-Trp-Leu-His-Ade) (TDO). The second screening was performed with the LacZ report gene ( yeast cells were grown in QDO medium containing X-alpha-gal). The interaction between HBV core protein and the protein obtained from positive colonies was further confirmed by repeating yeast-two hybrid. After plasmid DNA was extracted from blue colonies and sequenced, the results were analyzed by bioinformatic methods. RESULTS Eighteen colonies were obtained and sequenced, including hypermethylated in cancer 2 (3 colones), eukaryotic translation elongation factor 2 (2 colones), acetyl-coenzyme A synthetase 3 (1 colone), DNA polymerase gamma (1 colone), putative translation initiation factor (1 colone), chemokine (C-C motif) receptor 5 (1 colone), mitochondrial ribosomal protein L41 (1 colone), kyot binding protein genes (1 colone), RanBPM (1 colone), HBeAg-binding protein 3 (1 colone), programmed cell death 2 (1 colone). Four new genes with unknown function were identified. CONCLUSION Successful cloning of genes of HBV core protein interacting proteins in leukocytes may provide some new clues for studying the biological functions of HBV core protein.
Collapse
Affiliation(s)
- Shu-Mei Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Irshad M, Dhar I. Hepatitis C virus core protein: an update on its molecular biology, cellular functions and clinical implications. Med Princ Pract 2006; 15:405-16. [PMID: 17047346 DOI: 10.1159/000095485] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Accepted: 03/18/2006] [Indexed: 12/20/2022] Open
Abstract
The present review article is an update on various features of hepatitis C virus (HCV) core protein including its molecular biology, role in HCV replication, involvement in HCV pathogenesis, etiological role in hepatocellular carcinogenesis, significance in diagnosis and vaccination against HCV infection. Core protein is a structural protein of HCV virus and has only recently been characterized. It was found to play a major role in HCV-induced viral hepatitis. Although published information shows a lot about the clinical significance of HCV core protein, several studies are still needed to demonstrate its exact significance in viral biology and underlying HCV pathogenesis.
Collapse
Affiliation(s)
- M Irshad
- Clinical Biochemistry Division, Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, India.
| | | |
Collapse
|
48
|
Raghuraman S, Abraham P, Daniel HD, Ramakrishna BS, Sridharan G. Characterization of soluble FAS, FAS ligand and tumour necrosis factor-alpha in patients with chronic HCV infection. J Clin Virol 2005; 34:63-70. [PMID: 16087126 DOI: 10.1016/j.jcv.2005.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Revised: 01/03/2005] [Accepted: 01/11/2005] [Indexed: 12/12/2022]
Abstract
BACKGROUND There are limited reports on the role of the cell surface receptor Fas and its ligand molecule in mediating apoptosis during infection with the hepatitis C virus (HCV). OBJECTIVES The aims of this study were (1) to assess the susceptibility of the Fas antigen expressed on peripheral blood mononuclear cells to Fas ligand-induced-death in patients with chronic HCV infection and (2) to investigate the correlation between the plasma levels of soluble Fas (sFas), soluble Fas ligand (sFasL), tumour necrosis factor-alpha (TNF-alpha), alanine amino transferase (ALT), and HCV viral load. STUDY DESIGN The susceptibility of peripheral blood mononuclear cells from 17 subjects with chronic HCV infection to Fas ligand induced cell death was assessed using a water soluble tetrazolium assay. The plasma levels of associated markers such as sFas, sFasL, and TNF-alpha were quantified using immunoassays. ALT values were obtained from hospital records. Viral loads were quantified using a commercially available quantitative assay--the Amplicor Monitor (version 2.0). Controls for comparison included a group of healthy individuals and individuals infected with the human immunodeficiency virus 1. RESULTS The percentage of cell death induced in hepatitis C virus infected individuals was lower than that seen in the healthy control group. Patients infected with HCV had higher average values of sFas and TNF-alpha as compared to both control groups. Plasma levels of sFas in patients with chronic HCV infection showed significant positive correlations to ALT and TNF-alpha levels. TNF-alpha levels also showed a significant positive correlation with ALT levels. CONCLUSIONS PBMC in HCV infection exhibit decreased susceptibility to Fas ligand induced cell death. This may signify a means by which HCV escapes immune surveillance. This phenomenon merits further investigation. The strong correlations observed between plasma sFas, ALT and TNF-alpha suggest a potential role for these markers as an alternative to an invasive liver biopsy.
Collapse
Affiliation(s)
- Sukanya Raghuraman
- Department of Clinical Virology, Christian Medical College, Vellore 632004, TamilNadu, India.
| | | | | | | | | |
Collapse
|
49
|
Liao QJ, Ye LB, Timani KA, She YL, Yang XJ, Ye L, Wu ZH. Hepatitis C virus non-structural 5A protein can enhance full-length core protein-induced nuclear factor-kappaB activation. World J Gastroenterol 2005; 11:6433-9. [PMID: 16425412 PMCID: PMC4355782 DOI: 10.3748/wjg.v11.i41.6433] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2005] [Revised: 04/26/2005] [Accepted: 04/30/2005] [Indexed: 02/06/2023] Open
Abstract
AIM To study the effects of hepatitis C virus (HCV) core and non-structural 5A (NS5A) proteins on nuclear factor-kappaB (NF-kappaB) activity for understanding their biological function on chronic hepatitis caused by HCV infection. METHODS Luciferase assay was used to measure the activity of NF-kappaB in three different cell lines cotransfected with a series of deletion mutants of core protein alone or together with NS5A protein using pNF-kappaB-Luc as a reporter plasmid. Western blot and indirect immunofluorescence assays were used to confirm the expression of proteins and to detect their subcellular localization, respectively. Furthermore, Western blot was also used to detect the expression levels of NF-kappaB/p65, NF-kappaB/p50, and inhibitor kappaB-a (IkappaB-a). RESULTS The wild-type core protein (C191) and its mutant segments (C173 and C158) could activate NF-kappaB in Huh7 cells only and activation caused by (C191) could be enhanced by NS5A protein. Moreover, the full-length core protein and its different deletion mutants alone or together with NS5A protein did not enhance the expression level of NF-kappaB. The NF-kappaB activity was augmented due to the dissociation of NF-kappaB-IkappaB complex and the degradation of IkappaB-a. CONCLUSION NF-kappaB is the key transcription factor that can activate many genes that are involved in the cellular immune response and inflammation. Coexpression of the full-length core protein along with NS5A can enhance the NF-kappaB activation, and this activation may play a significant role in chronic liver diseases including hepatocellular carcinoma associated with HCV infection.
Collapse
Affiliation(s)
- Qing-Jiao Liao
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan 430072, Hubei Province, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Huang YP, Cheng J, Zhang SL, Wang L, Guo J, Liu Y, Yang Y, Zhang LY, Bai GQ, Gao XS, Ji D, Lin SM, Shao Q. Screening of hepatocyte proteins binding to F protein of hepatitis C virus by yeast two-hybrid system. World J Gastroenterol 2005; 11:5659-65. [PMID: 16237761 PMCID: PMC4481484 DOI: 10.3748/wjg.v11.i36.5659] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the biological function of F protein by yeast two-hybrid system.
METHODS: We constructed F protein bait plasmid by cloning the gene of F protein into pGBKT7, then recombinant plasmid DNA was transformed into yeast AH109 (a type). The transformed yeast AH109 was mated with yeast Y187 (α type) containing liver cDNA library plasmid in 2×YPDA medium. Diploid yeast was plated on synthetic dropout nutrient medium (SD/-Trp-Leu-His-Ade) containing X-α-gal for selection and screening. After extracting and sequencing plasmids from positive (blue) colonies, we underwent sequence analysis by bioinformatics.
RESULTS: Thirty-six colonies were selected and sequenced. Among them, 11 colonies were zymogen granule protein, 5 colonies were zinc finger protein, 4 colonies were zinc-α-2-glycoprotein, 1 colony was sialyltransferase, 1 colony was complement control protein factor I, 1 colony was vitronectin, and 2 colonies were new genes with unknown function.
CONCLUSION: The yeast two-hybrid system is an effective method for identifying hepatocyte proteins interacting with F protein of hepatitis C virus. F protein may bind to different proteins.
Collapse
Affiliation(s)
- Yan-Ping Huang
- Department of Pediatrics, the First Hospital of Xi'an Jiaotong University, Shaanxi Province, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|