1
|
Ou LL, Jiang JL, Guo ML, Wu JH, Zhong WW, He YH. Research progress on the roles of complement in liver injury. World J Hepatol 2025; 17:103839. [PMID: 40177195 PMCID: PMC11959660 DOI: 10.4254/wjh.v17.i3.103839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/29/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
The complement system is crucial for maintaining immunological homeostasis in the liver, playing a significant role in both innate and adaptive immune responses. Dysregulation of this system is closely linked to the pathogenesis of various liver diseases. Modulating the complement system can affect the progression of these conditions. To provide insights into treating liver injury by targeting the regulation of the complement system, we conducted a comprehensive search of major biomedical databases, including MEDLINE, PubMed, EMBASE, and Web of Science, to identify articles on complement and liver injury and reviewed the functions and mechanisms of the complement system in liver injury.
Collapse
Affiliation(s)
- Li-Li Ou
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Jin-Lian Jiang
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Man-Lu Guo
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Jin-Hua Wu
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Wei-Wei Zhong
- Department of Infectious Diseases, Jingmen Central Hospital, Jingmen Central Hospital Affiliated to Jingchu University of Technology, Jingmen 448000, Hubei Province, China
| | - Yi-Huai He
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, China.
| |
Collapse
|
2
|
Bellini N, Ye C, Ajibola O, Murooka TT, Lodge R, Cohen ÉA. Downregulation of miRNA-26a by HIV-1 Enhances CD59 Expression and Packaging, Impacting Virus Susceptibility to Antibody-Dependent Complement-Mediated Lysis. Viruses 2024; 16:1076. [PMID: 39066239 PMCID: PMC11281366 DOI: 10.3390/v16071076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/29/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
MicroRNAs (miRNAs) play important roles in the control of HIV-1 infection. Here, we performed RNA-seq profiling of miRNAs and mRNAs expressed in CD4+ T lymphocytes upon HIV-1 infection. Our results reveal significant alterations in miRNA and mRNA expression profiles in infected relative to uninfected cells. One of the miRNAs markedly downregulated in infected cells is miRNA-26a. Among the putative targets of miRNA-26a are CD59 receptor transcripts, which are significantly upregulated in infected CD4+ T cells. The addition of miRNA-26a mimics to CD4+ T cells reduces CD59 at both the mRNA and surface protein levels, validating CD59 as a miRNA-26a target. Consistent with the reported inhibitory role of CD59 in complement-mediated lysis (CML), knocking out CD59 in CD4+ T cells renders both HIV-1-infected cells and progeny virions more prone to antibody-dependent CML (ADCML). The addition of miRNA-26a mimics to infected cells leads to enhanced sensitivity of progeny virions to ADCML, a condition linked to a reduction in CD59 packaging into released virions. Lastly, HIV-1-mediated downregulation of miRNA-26a expression is shown to be dependent on integrated HIV-1 expression but does not involve viral accessory proteins. Overall, these results highlight a novel mechanism by which HIV-1 limits ADCML by upregulating CD59 expression via miRNA-26a downmodulation.
Collapse
Affiliation(s)
- Nicolas Bellini
- Laboratory of Human Retrovirology, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; (N.B.); (C.Y.); (R.L.)
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Chengyu Ye
- Laboratory of Human Retrovirology, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; (N.B.); (C.Y.); (R.L.)
| | - Oluwaseun Ajibola
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (O.A.); (T.T.M.)
| | - Thomas T. Murooka
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (O.A.); (T.T.M.)
| | - Robert Lodge
- Laboratory of Human Retrovirology, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; (N.B.); (C.Y.); (R.L.)
| | - Éric A. Cohen
- Laboratory of Human Retrovirology, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; (N.B.); (C.Y.); (R.L.)
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
3
|
Heggi MT, Nour El-Din HT, Morsy DI, Abdelaziz NI, Attia AS. Microbial evasion of the complement system: a continuous and evolving story. Front Immunol 2024; 14:1281096. [PMID: 38239357 PMCID: PMC10794618 DOI: 10.3389/fimmu.2023.1281096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/30/2023] [Indexed: 01/22/2024] Open
Abstract
The complement system is a fundamental part of the innate immune system that plays a key role in the battle of the human body against invading pathogens. Through its three pathways, represented by the classical, alternative, and lectin pathways, the complement system forms a tightly regulated network of soluble proteins, membrane-expressed receptors, and regulators with versatile protective and killing mechanisms. However, ingenious pathogens have developed strategies over the years to protect themselves from this complex part of the immune system. This review briefly discusses the sequence of the complement activation pathways. Then, we present a comprehensive updated overview of how the major four pathogenic groups, namely, bacteria, viruses, fungi, and parasites, control, modulate, and block the complement attacks at different steps of the complement cascade. We shed more light on the ability of those pathogens to deploy more than one mechanism to tackle the complement system in their path to establish infection within the human host.
Collapse
Affiliation(s)
- Mariam T. Heggi
- Clinical Pharmacy Undergraduate Program, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanzada T. Nour El-Din
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | | | | | - Ahmed S. Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
4
|
Bi W, Kraft A, Engelskircher S, Mischke J, Witte M, Klawonn F, van Ham M, Cornberg M, Wedemeyer H, Hengst J, Jänsch L. Proteomics reveals a global phenotypic shift of NK cells in HCV patients treated with direct-acting antivirals. Eur J Immunol 2023; 53:e2250291. [PMID: 37515498 DOI: 10.1002/eji.202250291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 07/31/2023]
Abstract
Chronic hepatitis C virus (HCV) infections compromise natural killer (NK)-cell immunity. Direct-acting antivirals (DAA) effectively eliminate HCV, but the long-term effects on NK cells in cured patients are debated. We conducted a proteomic study on CD56+ NK cells of chronic HCV-infected patients before and 1 year after DAA therapy. Donor-variation was observed in NK-cell proteomes of HCV-infected patients, with 46 dysregulated proteins restored after DAA therapy. However, 30% of the CD56+ NK-cell proteome remained altered 1 year post-therapy, indicating a phenotypic shift with low donor-variation. NK cells from virus-negative cured patients exhibited global regulation of RNA-processing and pathways related to "stimuli response", "chemokine signaling", and "cytotoxicity regulation". Proteomics identified downregulation of vesicle transport components (CD107a, COPI/II complexes) and altered receptor expression profiles, indicating an inhibited NK-cell phenotype. Yet, activated NK cells from HCV patients before and after therapy effectively upregulated IFN-γ and recruited CD107a. Conversely, reduced surface expression levels of Tim-3 and 2B4 were observed before and after therapy. In conclusion, this study reveals long-term effects on the CD56+ NK-cell compartment in convalescent HCV patients 1 year after therapy, with limited abundance of vesicle transport complexes and surface receptors, associated with a responsive NK-cell phenotype.
Collapse
Affiliation(s)
- Wenjie Bi
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, P. R. China
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anke Kraft
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Sophie Engelskircher
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Jasmin Mischke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Moana Witte
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Frank Klawonn
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Department of Computer Science, Ostfalia University, Wolfenbüttel, Germany
| | - Marco van Ham
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Heiner Wedemeyer
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany
| | - Julia Hengst
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Lothar Jänsch
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
5
|
Duda E. How much (evil) intelligence can be encoded by 30 kb? Biol Futur 2023:10.1007/s42977-023-00153-8. [PMID: 36752964 PMCID: PMC9907195 DOI: 10.1007/s42977-023-00153-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023]
Abstract
Genomes of most RNA viruses are rarely larger than the size of an average human gene (10-15 kb) and still code for a number of biologically active polypeptides that modify the immune system and metabolism of the host organism in an amazingly complex way. Prolonged coevolution developed tricks by which viruses can dodge many protective mechanisms of the host and lead to the formation of molecular mimicry patterns. Some viruses inhibit the interferon response, interfere with the membrane destroying effects of the activated complement cascade. They can replicate in cellular compartments formed by inner membranes of the cell hiding their characteristic features from diverse pattern recognition receptors. In many cases-and in this respect, the new coronavirus is a champion-they can exploit our own defensive mechanisms to cause serious harm, severe symptoms and frequently deadly disease.
Collapse
Affiliation(s)
- Ernő Duda
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary.
| |
Collapse
|
6
|
Tan Z, Fu S, Huang Y, Duan X, Zuo Y, Zhu X, Wang H, Wang J. HSPB8 is a Potential Prognostic Biomarker that Correlates With Immune Cell Infiltration in Bladder Cancer. Front Genet 2022; 13:804858. [PMID: 35330734 PMCID: PMC8940282 DOI: 10.3389/fgene.2022.804858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Heat shock protein B8 (HSPB8) is expressed in various cancers. However, the functional and clinicopathological significance of HSPB8 expression in bladder cancer (BC) remains unclear. The present study sought to elucidate the clinicopathological features and prognostic value of HSPB8 in BC. Methods: A BC RNA-seq data set was obtained from The Cancer Genome Atlas Urothelial Bladder Carcinoma (TCGA-BLCA) database, and the external validation dataset GSE130598 was downloaded from the GEO database. Samples in the TCGA-BLCA were categorized into two groups based on HSPB8 expression. Differentially expressed genes (DEGs) between the two groups were defined as HSPB8 co-expressed genes. Gene set enrichment analysis (GSEA), protein-protein interaction networks, and mRNA-microRNA (miRNA) interaction networks were generated to predict the function and interactions of genes that are co-expressed with HSPB8. Finally, we examined immune cell infiltration and constructed a survival prediction model for BC patients. Results: The expression level of HSBP8 has a significant difference between cancer samples and normal samples, and its diagnosis effect was validated by the ROC curve. 446 differential expressed genes between HSBP8 high-expression and HSBP8 low expression groups were identified. Gene enrichment analysis and GSEA analysis show that these differential gene functions are closely related to the occurrence and development of BC and the metabolic pathways of BC. The cancer-related pathways included Cytokine-cytokine receptor Interaction, Focal adhesion, and Proteoglycans in cancer. PPI and protein-coding gene-miRNA network visualized the landscape for these tightly bounded gene interactions. Immune cell infiltration shows that B cells, CD4+T cells, and CD8+T cells have strongly different infiltration levels between the HSBP8 high exp group and low exp group. The survival prediction model shows that HSBP8 has strong prognosis power in the BLCA cohort. Conclusion: Identifying DEGs may enhance understanding of BC development’s causes and molecular mechanisms. HSPB8 may play an essential role in BC progression and prognosis and serve as a potential biomarker for BC treatment.
Collapse
Affiliation(s)
- Zhiyong Tan
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
| | - Shi Fu
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
| | - Yinglong Huang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
| | - Xianzhong Duan
- Department of Urology, the Second People's Hospital of Baoshan, Baoshan, China
| | - Yigang Zuo
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
| | - Xiaorui Zhu
- Department of Urology, the Second People's Hospital of Baoshan, Baoshan, China
| | - Haifeng Wang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
| | - Jiansong Wang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
| |
Collapse
|
7
|
Malekshahi Z, Bernklau S, Schiela B, Koske I, Banki Z, Stiasny K, Harris CL, Würzner R, Stoiber H. Incorporation of CD55 into the Zika Viral Envelope Contributes to Its Stability against Human Complement. Viruses 2021; 13:v13030510. [PMID: 33808725 PMCID: PMC8003375 DOI: 10.3390/v13030510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 12/25/2022] Open
Abstract
The rapid spread of the virus in Latin America and the association of the infection with microcephaly in newborns or Guillain–Barré Syndrome in adults prompted the WHO to declare the Zika virus (ZIKV) epidemic to be an international public health emergency in 2016. As the virus was first discovered in monkeys and is spread not only by mosquitos but also from human to human, we investigated the stability to the human complement of ZIKV derived from mosquito (ZIKVInsect), monkey (ZIKVVero), or human cells (ZIKVA549 and ZIKVFibro), respectively. At a low serum concentration (10%), which refers to complement concentrations found on mucosal surfaces, the virus was relatively stable at 37 °C. At higher complement levels (up to 50% serum concentration), ZIKV titers differed significantly depending on the cell line used for the propagation of the virus. While the viral titer of ZIKVInsect decreased about two orders in magnitude, when incubated with human serum, the virus derived from human cells was more resistant to complement-mediated lysis (CML). By virus-capture assay and Western blots, the complement regulator protein CD55 was identified to be incorporated into the viral envelope. Blocking of CD55 by neutralizing Abs significantly increased the sensitivity to human complement. Taken together, these data indicate that the incorporation of CD55 from human cells contributes to the stability of ZIKV against complement-mediated virolysis.
Collapse
Affiliation(s)
- Zahra Malekshahi
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Sarah Bernklau
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Britta Schiela
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Iris Koske
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Zoltan Banki
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Claire L. Harris
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Reinhard Würzner
- Institute of Hygiene & Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (R.W.); (H.S.)
| | - Heribert Stoiber
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
- Correspondence: (R.W.); (H.S.)
| |
Collapse
|
8
|
Evaluation of Serum and Gene Expression of Galectin-4, Interleukin-27, and Complement-7 in Hepatitis C Virus-Infected Egyptian Patients. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8879758. [PMID: 33381596 PMCID: PMC7758134 DOI: 10.1155/2020/8879758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/10/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
Background Hepatitis C virus (HCV) is considered a major global public health problem. Recently, there are great advances in HCV therapy, but there are some limitations that are creating an urgent need for assessment of some cytokines that have a potent antiviral effect in the immune system and anti-inflammatory effects to provide a potential novel immunotherapeutic target in HCV infection. Objective This study was directed to assess the serum levels and gene expression levels of Galectin-4 (LEG4), Interleukin-27 (IL-27), and Complement-7 (C-7) and their correlation with the viral load in HCV infection. Subjects and Methods. This work was conducted on 80 subjects, Group 1 (n = 40) early detected HCV patients and Group 2 (n = 40) healthy controls. LEG4, IL-27, and C-7 were assessed at the protein levels by ELISA, and their gene expression was assessed by RT-qPCR. The viral load was measured by PCR. Results There were significant elevations in the mean levels of gene expression and serum levels of all studied parameters LEG4, IL-27, and C-7 in the HCV group compared to the control group. Significant negative correlations between the viral load and each of the serum proteins and gene expressions of both LEG4 and IL-27 in HCV patients were found. The gene expression levels of LEG4, IL-27, and C-7 were positively correlated with their corresponding serum proteins in HCV patients. Conclusion LEG4 and IL-27 showed significant negative correlations with the viral load, which could be an immune response to the control of the extent of hepatic inflammation, thus creating a potential novel immunotherapeutic approach in HCV infection for further studies or therapeutic clinical trials.
Collapse
|
9
|
Kumar NA, Kunnakkadan U, Thomas S, Johnson JB. In the Crosshairs: RNA Viruses OR Complement? Front Immunol 2020; 11:573583. [PMID: 33133089 PMCID: PMC7550403 DOI: 10.3389/fimmu.2020.573583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/24/2020] [Indexed: 12/02/2022] Open
Abstract
Complement, a part of the innate arm of the immune system, is integral to the frontline defense of the host against innumerable pathogens, which includes RNA viruses. Among the major groups of viruses, RNA viruses contribute significantly to the global mortality and morbidity index associated with viral infection. Despite multiple routes of entry adopted by these viruses, facing complement is inevitable. The initial interaction with complement and the nature of this interaction play an important role in determining host resistance versus susceptibility to the viral infection. Many RNA viruses are potent activators of complement, often resulting in virus neutralization. Yet, another facet of virus-induced activation is the exacerbation in pathogenesis contributing to the overall morbidity. The severity in disease and death associated with RNA virus infections shows a tip in the scale favoring viruses. Growing evidence suggest that like their DNA counterparts, RNA viruses have co-evolved to master ingenious strategies to remarkably restrict complement. Modulation of host genes involved in antiviral responses contributed prominently to the adoption of unique strategies to keep complement at bay, which included either down regulation of activation components (C3, C4) or up regulation of complement regulatory proteins. All this hints at a possible “hijacking” of the cross-talk mechanism of the host immune system. Enveloped RNA viruses have a selective advantage of not only modulating the host responses but also recruiting membrane-associated regulators of complement activation (RCAs). This review aims to highlight the significant progress in the understanding of RNA virus–complement interactions.
Collapse
Affiliation(s)
- Nisha Asok Kumar
- Viral Disease Biology, Department of Pathogen Biology, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, India.,Manipal Academy of Higher Education, Manipal, India
| | - Umerali Kunnakkadan
- Viral Disease Biology, Department of Pathogen Biology, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, India.,Department of Biotechnology, University of Kerala, Thiruvananthapuram, India
| | - Sabu Thomas
- Cholera and Biofilm Research Lab, Department of Pathogen Biology, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, India
| | - John Bernet Johnson
- Viral Disease Biology, Department of Pathogen Biology, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
10
|
Malekshahi Z, Schiela B, Bernklau S, Banki Z, Würzner R, Stoiber H. Interference of the Zika Virus E-Protein With the Membrane Attack Complex of the Complement System. Front Immunol 2020; 11:569549. [PMID: 33193347 PMCID: PMC7655927 DOI: 10.3389/fimmu.2020.569549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
The complement system has developed different strategies to clear infections by several effector mechanisms, such as opsonization, which supports phagocytosis, attracting immune cells by C3 and C5 cleavage products, or direct killing of pathogens by the formation of the membrane attack complex (MAC). As the Zika virus (ZIKV) activates the classical complement pathway and thus has to avoid clearance by the complement system, we analyzed putative viral escape mechanisms, which limit virolysis. We identified binding of the recombinant viral envelope E protein to components of the terminal pathway complement (C5b6, C7, C8, and C9) by ELISA. Western blot analyses revealed that ZIKV E protein interfered with the polymerization of C9, induced on cellular surfaces, either by purified terminal complement proteins or by normal human serum (NHS) as a source of the complement. Further, the hemolytic activity of NHS was significantly reduced in the presence of the recombinant E protein or entire viral particles. This data indicates that ZIKV reduces MAC formation and complement-mediated lysis by binding terminal complement proteins to the viral E protein.
Collapse
Affiliation(s)
- Zahra Malekshahi
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Britta Schiela
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Bernklau
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltan Banki
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinhard Würzner
- Institute of Hygiene & Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Heribert Stoiber
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Identification of potential mRNA panels for severe acute respiratory syndrome coronavirus 2 (COVID-19) diagnosis and treatment using microarray dataset and bioinformatics methods. 3 Biotech 2020; 10:422. [PMID: 33251083 PMCID: PMC7679428 DOI: 10.1007/s13205-020-02406-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/20/2020] [Indexed: 12/15/2022] Open
Abstract
The goal of the present investigation is to identify the differentially expressed genes (DEGs) between SARS-CoV-2 infected and normal control samples to investigate the molecular mechanisms of infection with SARS-CoV-2. The microarray data of the dataset E-MTAB-8871 were retrieved from the ArrayExpress database. Pathway and Gene Ontology (GO) enrichment study, protein–protein interaction (PPI) network, modules, target gene–miRNA regulatory network, and target gene–TF regulatory network have been performed. Subsequently, the key genes were validated using an analysis of the receiver operating characteristic (ROC) curve. In SARS-CoV-2 infection, a total of 324 DEGs (76 up- and 248 down-regulated genes) were identified and enriched in a number of associated SARS-CoV-2 infection pathways and GO terms. Hub and target genes such as TP53, HRAS, MAPK11, RELA, IKZF3, IFNAR2, SKI, TNFRSF13C, JAK1, TRAF6, KLRF2, CD1A were identified from PPI network, target gene–miRNA regulatory network, and target gene–TF regulatory network. Study of the ROC showed that ten genes (CCL5, IFNAR2, JAK2, MX1, STAT1, BID, CD55, CD80, HAL-B, and HLA-DMA) were substantially involved in SARS-CoV-2 patients. The present investigation identified key genes and pathways that deepen our understanding of the molecular mechanisms of SARS-CoV-2 infection, and could be used for SARS-CoV-2 infection as diagnostic and therapeutic biomarkers.
Collapse
|
12
|
Maloney BE, Perera KD, Saunders DRD, Shadipeni N, Fleming SD. Interactions of viruses and the humoral innate immune response. Clin Immunol 2020; 212:108351. [PMID: 32028020 DOI: 10.1016/j.clim.2020.108351] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/01/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022]
Abstract
The innate immune response is crucial for defense against virus infections where the complement system, coagulation cascade and natural antibodies play key roles. These immune components are interconnected in an intricate network and are tightly regulated to maintain homeostasis and avoid uncontrolled immune responses. Many viruses in turn have evolved to modulate these interactions through various strategies to evade innate immune activation. This review summarizes the current understanding on viral strategies to inhibit the activation of complement and coagulation cascades, evade natural antibody-mediated clearance and utilize complement regulatory mechanisms to their advantage.
Collapse
Affiliation(s)
- Bailey E Maloney
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Krishani Dinali Perera
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Danielle R D Saunders
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Naemi Shadipeni
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
13
|
Yu J, Murthy V, Liu SL. Relating GPI-Anchored Ly6 Proteins uPAR and CD59 to Viral Infection. Viruses 2019; 11:E1060. [PMID: 31739586 PMCID: PMC6893729 DOI: 10.3390/v11111060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022] Open
Abstract
The Ly6 (lymphocyte antigen-6)/uPAR (urokinase-type plasminogen activator receptor) superfamily protein is a group of molecules that share limited sequence homology but conserved three-fingered structures. Despite diverse cellular functions, such as in regulating host immunity, cell adhesion, and migration, the physiological roles of these factors in vivo remain poorly characterized. Notably, increasing research has focused on the interplays between Ly6/uPAR proteins and viral pathogens, the results of which have provided new insight into viral entry and virus-host interactions. While LY6E (lymphocyte antigen 6 family member E), one key member of the Ly6E/uPAR-family proteins, has been extensively studied, other members have not been well characterized. Here, we summarize current knowledge of Ly6/uPAR proteins related to viral infection, with a focus on uPAR and CD59. Our goal is to provide an up-to-date view of the Ly6/uPAR-family proteins and associated virus-host interaction and viral pathogenesis.
Collapse
Affiliation(s)
- Jingyou Yu
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Vaibhav Murthy
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Shan-Lu Liu
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
14
|
Abstract
A prominent role for complement has been identified in the linkage of innate and adaptive immunity. The liver is the main source of complement and hepatocytes are the primary sites for synthesis of complement components in vivo. We have discovered that hepatitis C virus (HCV) impairs C4 and C3 synthesis. Liver damage may diminish capacity of complement synthesis in patients. However, we observed that the changes in measured complement components in chronically HCV infected patients do not correlate with liver fibrosis or rheumatoid factor present in the blood, serum albumin, or alkaline phosphatase levels. Complement component C3 is of critical importance in B cell activation and T cell-dependent antibody responses. C3 activity is required for optimal expansion of CD8+T cells during a systemic viral infection. Deficiencies in complement may predispose patients to infections via ineffective opsonization, and defects in lytic activity via membrane attack complex. Interestingly, C9 is significantly reduced at the mRNA level in chronically HCV infected liver biopsy specimens, while many hepatocyte derived complement components (C6, C8, Factor B, MASP1, and MBL) and unrelated genes remain mostly unaffected. This implies an HCV specific effect, not a global effect from liver disease.
Collapse
Affiliation(s)
- Young-Chan Kwon
- Department of Internal Medicine, Saint Louis University, St. Louis, MO, USA
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, USA
- Institut Pasteur Korea, Daejeon, Republic of Korea
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, St. Louis, MO, USA.
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, USA.
| |
Collapse
|
15
|
El-Shamy A, Branch AD, Schiano TD, Gorevic PD. The Complement System and C1q in Chronic Hepatitis C Virus Infection and Mixed Cryoglobulinemia. Front Immunol 2018; 9:1001. [PMID: 29910796 PMCID: PMC5992393 DOI: 10.3389/fimmu.2018.01001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022] Open
Abstract
The complement system bridges innate and adaptive immunity against microbial infections, with viral infection being a major trigger. Activation of the classical, alternative, and lectin pathways have been reported in chronic hepatitis C virus (HCV) infection and/or cryoglobulinemia. HCV infection leads to dysregulation of complement-mediated immune responses. Clinical and experimental evidence support involvement of complement in intra- and extrahepatic manifestations of HCV infection, such as liver fibrosis and type II cryoglobulinemia. In this review, we summarize studies that have investigated the interplay between HCV and the complement system to establish chronic infection and autoimmunity, as well as the association between HCV pathogenesis and abnormal complement profiles. Several unanswered questions are highlighted which suggest additional informative lines of investigation.
Collapse
Affiliation(s)
- Ahmed El-Shamy
- Division of Liver Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Pharmaceutical and Biological Sciences, California Northstate University, Elk Grove, CA, United States
| | - Andrea D Branch
- Division of Liver Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Thomas D Schiano
- Division of Liver Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Peter D Gorevic
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
16
|
Silva AA, Catarino SJ, Boldt ABW, Pedroso MLA, Beltrame MH, Messias-Reason IJ. Effects of MASP2 haplotypes and MASP-2 levels in hepatitis C-infected patients. Int J Immunogenet 2018; 45:118-127. [PMID: 29675993 DOI: 10.1111/iji.12371] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 02/15/2018] [Accepted: 03/22/2018] [Indexed: 01/01/2023]
Abstract
Mannan-binding lectin (MBL) and MBL-associated serine protease 2 (MASP-2) are components of the lectin pathway, which activate the complement system after binding to the HCV structural proteins E1 and E2. We haplotyped 11 MASP2 polymorphisms in 103 HCV patients and 205 controls and measured MASP-2 levels in 67 HCV patients and 77 controls to better understand the role of MASP-2 in hepatitis C susceptibility and disease severity according to viral genotype and fibrosis levels. The haplotype block MASP2*ARDP was associated with protection against HCV infection (OR = 0.49, p = .044) and lower MASP-2 levels in controls (p = .021), while haplotype block AGTDVRC was significantly increased in patients (OR = 7.58, p = .003). MASP-2 levels were lower in patients than in controls (p < .001) and in patients with viral genotype 1 or 4 (poor responders to treatment) than genotype 3 (p = .022) and correlated inversely with the levels of alkaline phosphatase, especially in individuals with fibrosis 3 or 4 (R = -.7, p = .005). MASP2 gene polymorphisms modulate basal gene expression, which may influence the quality of complement response against HCV. MASP-2 levels decrease during chronic disease, independently of MASP2 genotypes, most probably due to consumption and attenuation mechanisms of viral origin and by the reduced liver function, the site of MASP-2 production.
Collapse
Affiliation(s)
- Amanda A Silva
- Departamento de Patologia Médica, Hospital de Clínicas, Laboratório de Imunopatologia Molecular, Universidade Federal do Paraná, Curitiba, Brazil
| | - Sandra J Catarino
- Departamento de Patologia Médica, Hospital de Clínicas, Laboratório de Imunopatologia Molecular, Universidade Federal do Paraná, Curitiba, Brazil
| | - Angelica B W Boldt
- Laboratório de Genética Molecular Humana, Universidade Federal do Paraná, Curitiba, Brazil
| | - Maria Lucia A Pedroso
- Departamento de Clínica Médica, Hospital de Clínicas, Serviço de Hepatologia, Universidade Federal do Paraná, Curitiba, Brazil
| | - Marcia H Beltrame
- Laboratório de Genética Molecular Humana, Universidade Federal do Paraná, Curitiba, Brazil
| | - Iara J Messias-Reason
- Departamento de Patologia Médica, Hospital de Clínicas, Laboratório de Imunopatologia Molecular, Universidade Federal do Paraná, Curitiba, Brazil
| |
Collapse
|
17
|
Wei Y, Ji Y, Guo H, Zhi X, Han S, Zhang Y, Gao Y, Chang Y, Yan D, Li K, Liu DX, Sun S. CD59 association with infectious bronchitis virus particles protects against antibody-dependent complement-mediated lysis. J Gen Virol 2017; 98:2725-2730. [PMID: 29068273 DOI: 10.1099/jgv.0.000962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
CD59 protein functions as a negative regulator of the terminal pathway of the complement system by binding to the C8/C9 factors. To date, little is known about the role of CD59 in coronavirus infectious bronchitis virus (IBV) infection. In this study, we discovered that CD59 was downregulated in IBV-infected cells and was associated with IBV virions. This association protected IBV particles from antibody-dependent complement-mediated lysis. IBV titres in the supernatant were significantly increased when CD59 proteins were overexpressed in cells followed by IBV infection, and this observation was further supported by knockdown or cleavage of CD59. Because no considerable change in IBV N protein and viral RNA levels was detected in total cell lysates prepared from the overexpression, knockdown or cleavage of CD59 groups, our data indicated that CD59 was involved in IBV particle release and that IBV had evolved a mechanism to utilize CD59 to evade complement-mediated destruction.
Collapse
Affiliation(s)
- Yanquan Wei
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Yanhong Ji
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Xiaoying Zhi
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Shichong Han
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Yun Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Yuan Gao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Yanyan Chang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Dan Yan
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Kangyu Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Ding Xiang Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China.,Guangdong Province Key Laboratory Microbial Signals and Disease Co, and Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, Guangdong 510642, PR China
| | - Shiqi Sun
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| |
Collapse
|
18
|
Hill DL, Schofield L, Wilson DW. IgG opsonization of merozoites: multiple immune mechanisms for malaria vaccine development. Int J Parasitol 2017; 47:585-595. [PMID: 28668325 DOI: 10.1016/j.ijpara.2017.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/12/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Global eradication of the human-infecting malaria parasite Plasmodium falciparum, the major cause of malaria mortality, is unlikely to be achieved without an effective vaccine. However, our limited understanding of how protective immune responses target malaria parasites in humans, and how to best elicit these immune responses through vaccination, has hampered vaccine development. The red blood cell invading stage of the parasite lifecycle (merozoite) displays antigens that are attractive vaccine candidates as they are accessible to antibodies and raise high antibody titres in naturally immune individuals. The number of merozoite antigens that elicit an immune response, and their structural and functional diversity, has led to a large number of lead antigens being pursued as vaccine candidates. Despite being seemingly spoilt for choice in terms of vaccine candidates, there is still a lack of consensus on exactly how merozoite antibodies reduce parasitemia and malaria disease. In this review we describe the various immune mechanisms that can result from IgG opsonization of merozoites, and highlight recent developments that support a role for these functional antibodies in naturally acquired and vaccine-induced immunity.
Collapse
Affiliation(s)
- Danika L Hill
- Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom; The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia.
| | - Louis Schofield
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia; Burnet Institute, 85 Commercial Road, Melbourne 3004, Victoria, Australia.
| |
Collapse
|
19
|
Agrawal P, Nawadkar R, Ojha H, Kumar J, Sahu A. Complement Evasion Strategies of Viruses: An Overview. Front Microbiol 2017; 8:1117. [PMID: 28670306 PMCID: PMC5472698 DOI: 10.3389/fmicb.2017.01117] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/31/2017] [Indexed: 12/11/2022] Open
Abstract
Being a major first line of immune defense, the complement system keeps a constant vigil against viruses. Its ability to recognize large panoply of viruses and virus-infected cells, and trigger the effector pathways, results in neutralization of viruses and killing of the infected cells. This selection pressure exerted by complement on viruses has made them evolve a multitude of countermeasures. These include targeting the recognition molecules for the avoidance of detection, targeting key enzymes and complexes of the complement pathways like C3 convertases and C5b-9 formation - either by encoding complement regulators or by recruiting membrane-bound and soluble host complement regulators, cleaving complement proteins by encoding protease, and inhibiting the synthesis of complement proteins. Additionally, viruses also exploit the complement system for their own benefit. For example, they use complement receptors as well as membrane regulators for cellular entry as well as their spread. Here, we provide an overview on the complement subversion mechanisms adopted by the members of various viral families including Poxviridae, Herpesviridae, Adenoviridae, Flaviviridae, Retroviridae, Picornaviridae, Astroviridae, Togaviridae, Orthomyxoviridae and Paramyxoviridae.
Collapse
Affiliation(s)
- Palak Agrawal
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Renuka Nawadkar
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Hina Ojha
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Jitendra Kumar
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Arvind Sahu
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| |
Collapse
|
20
|
Salviano-Silva A, Petzl-Erler ML, Boldt ABW. CD59 polymorphisms are associated with gene expression and different sexual susceptibility to pemphigus foliaceus. Autoimmunity 2017; 50:377-385. [DOI: 10.1080/08916934.2017.1329830] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Amanda Salviano-Silva
- Laboratory of Human Molecular Genetics, Department of Genetics, Universidade Federal do Paraná, Curitiba, Brazil
| | - Maria Luiza Petzl-Erler
- Laboratory of Human Molecular Genetics, Department of Genetics, Universidade Federal do Paraná, Curitiba, Brazil
| | - Angelica Beate Winter Boldt
- Laboratory of Human Molecular Genetics, Department of Genetics, Universidade Federal do Paraná, Curitiba, Brazil
- Laboratory of Molecular Immunopathology, Department of Clinical Pathology, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, Brazil
| |
Collapse
|
21
|
Ludwig A, Nguyen TH, Leong D, Ravi LI, Tan BH, Sandin S, Sugrue RJ. Caveolae provide a specialized membrane environment for respiratory syncytial virus assembly. J Cell Sci 2017; 130:1037-1050. [PMID: 28154158 PMCID: PMC5358342 DOI: 10.1242/jcs.198853] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/26/2017] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is an enveloped virus that assembles into filamentous virus particles on the surface of infected cells. Morphogenesis of RSV is dependent upon cholesterol-rich (lipid raft) membrane microdomains, but the specific role of individual raft molecules in RSV assembly is not well defined. Here, we show that RSV morphogenesis occurs within caveolar membranes and that both caveolin-1 and cavin-1 (also known as PTRF), the two major structural and functional components of caveolae, are actively recruited to and incorporated into the RSV envelope. The recruitment of caveolae occurred just prior to the initiation of RSV filament assembly, and was dependent upon an intact actin network as well as a direct physical interaction between caveolin-1 and the viral G protein. Moreover, cavin-1 protein levels were significantly increased in RSV-infected cells, leading to a virus-induced change in the stoichiometry and biophysical properties of the caveolar coat complex. Our data indicate that RSV exploits caveolae for its assembly, and we propose that the incorporation of caveolae into the virus contributes to defining the biological properties of the RSV envelope.
Collapse
Affiliation(s)
- Alexander Ludwig
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Tra Huong Nguyen
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Daniel Leong
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510
| | - Laxmi Iyer Ravi
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Boon Huan Tan
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510
| | - Sara Sandin
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| |
Collapse
|
22
|
Wang RY, Bare P, De Giorgi V, Matsuura K, Salam KA, Grandinetti T, Schechterly C, Alter HJ. Preferential association of hepatitis C virus with CD19 + B cells is mediated by complement system. Hepatology 2016; 64:1900-1910. [PMID: 27641977 PMCID: PMC5115962 DOI: 10.1002/hep.28842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/08/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023]
Abstract
Extrahepatic disease manifestations are common in chronic hepatitis C virus (HCV) infection. The mechanism of HCV-related lymphoproliferative disorders is not fully understood. Recent studies have found that HCV in peripheral blood mononuclear cells from chronically infected patients is mainly associated with cluster of differentiation 19-positive (CD19+ ) B cells. To further elucidate this preferential association of HCV with B cells, we used in vitro cultured virus and uninfected peripheral blood mononuclear cells from healthy blood donors to investigate the necessary serum components that activate the binding of HCV to B cells. First, we found that the active serum components were present not only in HCV carriers but also in HCV recovered patients and HCV-negative, healthy blood donors and that the serum components were heat-labile. Second, the preferential binding activity of HCV to B cells could be blocked by anti-complement C3 antibodies. In experiments with complement-depleted serum and purified complement proteins, we demonstrated that complement proteins C1, C2, and C3 were required to activate such binding activity. Complement protein C4 was partially involved in this process. Third, using antibodies against cell surface markers, we showed that the binding complex mainly involved CD21 (complement receptor 2), CD19, CD20, and CD81; CD35 (complement receptor 1) was involved but had lower binding activity. Fourth, both anti-CD21 and anti-CD35 antibodies could block the binding of patient-derived HCV to B cells. Fifth, complement also mediated HCV binding to Raji cells, a cultured B-cell line derived from Burkitt's lymphoma. CONCLUSION In chronic HCV infection, the preferential association of HCV with B cells is mediated by the complement system, mainly through complement receptor 2 (CD21), in conjunction with the CD19 and CD81 complex. (Hepatology 2016;64:1900-1910).
Collapse
Affiliation(s)
- Richard Y. Wang
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Patricia Bare
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
- Instituto de Investigaciones Hematológicas, Instituto de Medicina Experimental, CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Valeria De Giorgi
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Kentaro Matsuura
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazi Abdus Salam
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Teresa Grandinetti
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Cathy Schechterly
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Harvey J. Alter
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| |
Collapse
|
23
|
Abou-El-Hassan H, Zaraket H. Viral-derived complement inhibitors: current status and potential role in immunomodulation. Exp Biol Med (Maywood) 2016; 242:397-410. [PMID: 27798122 DOI: 10.1177/1535370216675772] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The complement system is one of the body's major innate immune defense mechanisms in vertebrates. Its function is to detect foreign bodies and promote their elimination through opsonisation or lysis. Complement proteins play an important role in the immunopathogenesis of several disorders. However, excessive complement activation does not confer more protection but instead leads to several autoimmune and inflammatory diseases. With inappropriate activation of the complement system, activated complement proteins and glycoproteins may damage both healthy and diseased tissues. Development of complement inhibitors represents an effective approach in controlling dysregulated complement activity and reducing disease severity, yet few studies have investigated the nature and role of novel complement inhibitory proteins of viral origin. Viral complement inhibitors have important implications in understanding the importance of complement inhibition and their role as a promising novel therapeutic approach in diseases caused by dysregulated complement function. In this review, we discuss the role and importance of complement inhibitors derived from several viruses in the scope of human inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- 1 Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon.,2 Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Zaraket
- 2 Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,3 Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
24
|
Amet T, Lan J, Shepherd N, Yang K, Byrd D, Xing Y, Yu Q. Glycosylphosphatidylinositol Anchor Deficiency Attenuates the Production of Infectious HIV-1 and Renders Virions Sensitive to Complement Attack. AIDS Res Hum Retroviruses 2016; 32:1100-1112. [PMID: 27231035 PMCID: PMC5067833 DOI: 10.1089/aid.2016.0046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) escapes complement-mediated lysis (CML) by incorporating host regulators of complement activation (RCA) into its envelope. CD59, a key member of RCA, is incorporated into HIV-1 virions at levels that protect against CML. Since CD59 is a glycosylphosphatidylinositol-anchored protein (GPI-AP), we used GPI anchor-deficient Jurkat cells (Jurkat-7) that express intracellular CD59, but not surface CD59, to study the molecular mechanisms underlying CD59 incorporation into HIV-1 virions and the role of host proteins in virus replication. Compared to Jurkat cells, Jurkat-7 cells were less supportive to HIV-1 replication and more sensitive to CML. Jurkat-7 cells exhibited similar capacities of HIV-1 binding and entry to Jurkat cells, but were less supportive to viral RNA and DNA biosynthesis as infected Jurkat-7 cells produced reduced amounts of HIV-1 RNA and DNA. HIV-1 virions produced from Jurkat-7 cells were CD59 negative, suggesting that viral particles acquire CD59, and probably other host proteins, from the cell membrane rather than intracellular compartments. As a result, CD59-negative virions were sensitive to CML. Strikingly, these virions exhibited reduced activity of virus binding and were less infectious, implicating that GPI-APs may be also important in ensuring the integrity of HIV-1 particles. Transient expression of the PIG-A gene restored CD59 expression on the surface of Jurkat-7 cells. After HIV-1 infection, the restored CD59 was colocalized with viral envelope glycoprotein gp120/gp41 within lipid rafts, which is identical to that on infected Jurkat cells. Thus, HIV-1 virions acquire RCA from the cell surface, likely lipid rafts, to escape CML and ensure viral infectivity.
Collapse
Affiliation(s)
- Tohti Amet
- Department of Microbiology and Immunology, Indiana Center for AIDS Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jie Lan
- Department of Microbiology and Immunology, Indiana Center for AIDS Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nicole Shepherd
- Department of Microbiology and Immunology, Indiana Center for AIDS Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kai Yang
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou, China
| | - Daniel Byrd
- Department of Microbiology and Immunology, Indiana Center for AIDS Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yanyan Xing
- Department of Microbiology and Immunology, Indiana Center for AIDS Research, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Pathophysiology, Medical College of Jinan University, Guangzhou, China
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana Center for AIDS Research, Indiana University School of Medicine, Indianapolis, Indiana
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou, China
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
25
|
Kwon YC, Kim H, Meyer K, Di Bisceglie AM, Ray R. Distinct CD55 Isoform Synthesis and Inhibition of Complement-Dependent Cytolysis by Hepatitis C Virus. THE JOURNAL OF IMMUNOLOGY 2016; 197:1127-36. [PMID: 27357152 DOI: 10.4049/jimmunol.1600631] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/02/2016] [Indexed: 01/31/2023]
Abstract
CD55/DAF, one of the regulators of complement activation, is known to limit excess complement activation on the host cell surface by accelerating the decay of C3 convertase. We reported previously that hepatitis C virus (HCV) infection or virus core protein expression upregulates CD55 expression. CD55 associates with HCV particles, potentially protecting HCV from lysis in circulation. An increase in CD55 on the surface of HCV-infected cells may inhibit complement-mediated cell killing. In this study, we show that Abs against cancer cell surface proteins induce complement-dependent cytolysis or Ab-dependent cell-mediated cytotoxicity of immortalized human hepatocytes in the presence of CD55-blocking Ab. CD55 has a secreted isoform (sCD55) that is generated by alternative splicing. We observed that sCD55 is induced in HCV-infected or HCV replicon-harboring cells, as well as in liver biopsy samples from chronically HCV-infected patients. Conditioned medium from HCV-infected hepatoma cells (Huh7.5 cells) or immortalized human hepatocytes inhibited C3 convertase activity and complement-dependent cytolysis of sheep blood erythrocytes. Chronically HCV-infected patient sera inhibited C3 convertase activity, further implicating HCV-specific impairment of complement function in infected humans. CD55-blocking Ab inhibited erythrocyte lysis by conditioned medium, suggesting that CD55/sCD55 impairs convertase activity. Together, our data show that HCV infection induces sCD55 expression in HCV-infected cell culture-conditioned medium and inhibits C3 convertase activity. This may have implications for modulating complement-mediated immune function in the microenvironment and on HCV-harboring cells.
Collapse
Affiliation(s)
- Young-Chan Kwon
- Department of Internal Medicine, Saint Louis University, St. Louis, MO 63104; and
| | - Hangeun Kim
- Department of Internal Medicine, Saint Louis University, St. Louis, MO 63104; and
| | - Keith Meyer
- Department of Internal Medicine, Saint Louis University, St. Louis, MO 63104; and
| | | | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, St. Louis, MO 63104; and Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63140
| |
Collapse
|
26
|
Ibáñez-Escribano A, Nogal-Ruiz JJ, Pérez-Serrano J, Gómez-Barrio A, Escario JA, Alderete J. Sequestration of host-CD59 as potential immune evasion strategy of Trichomonas vaginalis. Acta Trop 2015; 149:1-7. [PMID: 25976413 DOI: 10.1016/j.actatropica.2015.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/20/2015] [Accepted: 05/04/2015] [Indexed: 12/26/2022]
Abstract
Trichomonas vaginalis is known to evade complement-mediated lysis. Because the genome of T. vaginalis does not possess DNA sequence with homology to human protectin (CD59), a complement lysis restricting factor, we tested the hypothesis that host CD59 acquisition by T. vaginalis organisms mediates resistance to complement killing. This hypothesis was based on the fact that trichomonads are known to associate with host proteins. No CD59 was detected on the surface of T. vaginalis grown in serum-based medium using as probe anti-CD59 monoclonal antibody (MAb). We, therefore, infected mice intraperitoneally with live T. vaginalis, and trichomonads harvested from ascites were tested for binding of CD59. Immunofluorescence showed that parasites had surface CD59. Furthermore, as mouse erythrocytes (RBCs) possess membrane-associated CD59, and trichomonads use RBCs as a nutrient source, organisms were co-cultured with murine RBCs for one week. Parasites were shown to have detectable surface CD59. Importantly, live T. vaginalis with bound CD59 were compared with batch-grown parasites without surface-associated CD59 for sensitivity to complement in human serum. Trichomonads without surface-bound CD59 had a higher level of killing by complement than did parasites with surface CD59. These data show that host CD59 acquired onto the surface by live T. vaginalis may be an alternative mechanism for complement evasion. We describe a novel strategy by T. vaginalis consistent with host protein procurement by this parasite to evade the lytic action of complement.
Collapse
|
27
|
Gan Z, Wang B, Zhou W, Lu Y, Zhu W, Tang J, Jian J, Wu Z. Molecular and functional characterization of CD59 from Nile tilapia (Oreochromis niloticus) involved in the immune response to Streptococcus agalactiae. FISH & SHELLFISH IMMUNOLOGY 2015; 44:50-59. [PMID: 25661843 DOI: 10.1016/j.fsi.2015.01.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 06/04/2023]
Abstract
CD59, the major inhibitor of membrane attack complex, plays a crucial role in regulation of complement activation. In this paper, a CD59 gene of Nile tilapia, Oreochromis niloticus (designated as On-CD59) was cloned and its expression pattern under the stimulation of Streptococcus agalactiae was investigated. Sequence analysis showed main structural features required for complement-inhibitory activity were detected in the deduced amino acid sequence of On-CD59. In healthy Nile tilapia, the On-CD59 transcripts could be detected in all the examined tissues, with the most abundant expression in the brain. When immunized with inactivated S. agalactiae, there was a clear time-dependent expression pattern of On-CD59 in the skin, brain, head kidney, thymus and spleen, with quite different kinetic expressions. The assays for the complement-inhibitory activity suggested that recombinant On-CD59 protein had a species-selective inhibition of complement. Moreover, our works showed that recombinant On-CD59 protein may possess both binding activities to PGN and LTA and inhibiting activity of S. agalactiae. These findings indicated that On-CD59 may play important roles in the immune response to S. agalactiae in Nile tilapia.
Collapse
Affiliation(s)
- Zhen Gan
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Bei Wang
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Wei Zhou
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Yishan Lu
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China.
| | - Weiwei Zhu
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Jufen Tang
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - JiChang Jian
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Zaohe Wu
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| |
Collapse
|
28
|
Kwon YC, Ray RB, Ray R. Hepatitis C virus infection: establishment of chronicity and liver disease progression. EXCLI JOURNAL 2014; 13:977-96. [PMID: 26417315 PMCID: PMC4464452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/14/2014] [Indexed: 11/16/2022]
Abstract
Hepatitis C virus (HCV) often causes persistent infection, and is an important factor in the etiology of fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). There are no preventive or therapeutic vaccines available against HCV. Treatment strategies of HCV infection are likely to improve with recently discovered direct antiviral agents (DAAs). However, a proportion of patients still progress to liver failure and/or HCC despite having been cured of the infection. Thus, there is a need for early diagnosis and therapeutic modalities for HCV related end stage liver disease prevention. HCV genome does not integrate into its host genome, and has a predominantly cytoplasmic life cycle. Therefore, HCV mediated liver disease progression appears to involve indirect mechanisms from persistent infection of hepatocytes. Studying the underlying mechanisms of HCV mediated evasion of immune responses and liver disease progression is challenging due to the lack of a naturally susceptible small animal model. We and other investigators have used a number of experimental systems to investigate the mechanisms for establishment of chronic HCV infection and liver disease progression. HCV infection modulates immune systems. Further, HCV infection of primary human hepatocytes promotes growth, induces phenotypic changes, modulates epithelial mesenchymal transition (EMT) related genes, and generates tumor initiating stem-like cells (TISCs). HCV infection also modulates microRNAs (miRNAs), and influences growth by overriding normal death progression of primary human hepatocytes for disease pathogenesis. Understanding these ob-servations at the molecular level should aid in developing strategies for additional effective therapies against HCV mediated liver disease progression.
Collapse
Affiliation(s)
- Young-Chan Kwon
- Department of Internal Medicine, Saint Louis University, Missouri
| | - Ratna B. Ray
- Department of Pathology, Saint Louis University, Missouri
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, Missouri,Department of Molecular Microbiology & Immunology, Saint Louis University, Missouri,*To whom correspondence should be addressed: Ranjit Ray, Division of Infectious Diseases, Allergy & Immunology, Edward A. Doisy Research Center, 1100 S. Grand Blvd, 8th Floor, St. Louis, MO 63104, USA, E-mail:
| |
Collapse
|
29
|
Lan J, Yang K, Byrd D, Hu N, Amet T, Shepherd N, Desai M, Gao J, Gupta S, Sun Y, Yu Q. Provirus activation plus CD59 blockage triggers antibody-dependent complement-mediated lysis of latently HIV-1-infected cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:3577-89. [PMID: 25149467 DOI: 10.4049/jimmunol.1303030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Latently HIV-1-infected cells are recognized as the last barrier toward viral eradication and cure. To purge these cells, we combined a provirus stimulant with a blocker of human CD59, a key member of the regulators of complement activation, to trigger Ab-dependent complement-mediated lysis. Provirus stimulants including prostratin and histone deacetylase inhibitors such as romidepsin and suberoylanilide hydroxamic acid activated proviruses in the latently HIV-1-infected T cell line ACH-2 as virion production and viral protein expression on the cell surface were induced. Romidepsin was the most attractive provirus stimulant as it effectively activated proviruses at nanomolar concentrations that can be achieved clinically. Antiretroviral drugs including two protease inhibitors (atazanavir and darunavir) and an RT inhibitor (emtricitabine) did not affect the activity of provirus stimulants in the activation of proviruses. However, saquinavir (a protease inhibitor) markedly suppressed virus production, although it did not affect the percentage of cells expressing viral Env on the cell surface. Provirus-activated ACH-2 cells expressed HIV-1 Env that colocalized with CD59 in lipid rafts on the cell surface, facilitating direct interaction between them. Blockage of CD59 rendered provirus-activated ACH-2 cells and primary human CD4(+) T cells that were latently infected with HIV-1 sensitive to Ab-dependent complement-mediated lysis by anti-HIV-1 polyclonal Abs or plasma from HIV-1-infected patients. Therefore, a combination of provirus stimulants with regulators of complement activation blockers represents a novel approach to eliminate HIV-1.
Collapse
Affiliation(s)
- Jie Lan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202; Center for AIDS Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Kai Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202; Center for AIDS Research, Indiana University School of Medicine, Indianapolis, IN 46202; Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Wenzhou Medical College, Wenzhou 325035, China
| | - Daniel Byrd
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202; Center for AIDS Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Ningjie Hu
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Wenzhou Medical College, Wenzhou 325035, China
| | - Tohti Amet
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202; Center for AIDS Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Nicole Shepherd
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202; Center for AIDS Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Mona Desai
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202; and
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Wenzhou Medical College, Wenzhou 325035, China
| | - Samir Gupta
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202; and
| | - Yongtao Sun
- Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202; Center for AIDS Research, Indiana University School of Medicine, Indianapolis, IN 46202; Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202; and
| |
Collapse
|
30
|
Moerdyk-Schauwecker M, Hwang SI, Grdzelishvili VZ. Cellular proteins associated with the interior and exterior of vesicular stomatitis virus virions. PLoS One 2014; 9:e104688. [PMID: 25105980 PMCID: PMC4126742 DOI: 10.1371/journal.pone.0104688] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/15/2014] [Indexed: 01/18/2023] Open
Abstract
Virus particles (virions) often contain not only virus-encoded but also host-encoded proteins. Some of these host proteins are enclosed within the virion structure, while others, in the case of enveloped viruses, are embedded in the host-derived membrane. While many of these host protein incorporations are likely accidental, some may play a role in virus infectivity, replication and/or immunoreactivity in the next host. Host protein incorporations may be especially important in therapeutic applications where large numbers of virus particles are administered. Vesicular stomatitis virus (VSV) is the prototypic rhabdovirus and a candidate vaccine, gene therapy and oncolytic vector. Using mass spectrometry, we previously examined cell type dependent host protein content of VSV virions using intact (“whole”) virions purified from three cell lines originating from different species. Here we aimed to determine the localization of host proteins within the VSV virions by analyzing: i) whole VSV virions; and ii) whole VSV virions treated with Proteinase K to remove all proteins outside the viral envelope. A total of 257 proteins were identified, with 181 identified in whole virions and 183 identified in Proteinase K treated virions. Most of these proteins have not been previously shown to be associated with VSV. Functional enrichment analysis indicated the most overrepresented categories were proteins associated with vesicles, vesicle-mediated transport and protein localization. Using western blotting, the presence of several host proteins, including some not previously shown in association with VSV (such as Yes1, Prl1 and Ddx3y), was confirmed and their relative quantities in various virion fractions determined. Our study provides a valuable inventory of virion-associated host proteins for further investigation of their roles in the replication cycle, pathogenesis and immunoreactivity of VSV.
Collapse
Affiliation(s)
- Megan Moerdyk-Schauwecker
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
- Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Sun-Il Hwang
- Proteomics Laboratory for Clinical and Translational Research, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
| | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
- Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
31
|
Ojha H, Panwar HS, Gorham RD, Morikis D, Sahu A. Viral regulators of complement activation: structure, function and evolution. Mol Immunol 2014; 61:89-99. [PMID: 24976595 DOI: 10.1016/j.molimm.2014.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 05/30/2014] [Accepted: 06/01/2014] [Indexed: 11/25/2022]
Abstract
The complement system surveillance in the host is effective in controlling viral propagation. Consequently, to subvert this effector mechanism, viruses have developed a series of adaptations. One among these is encoding mimics of host regulators of complement activation (RCA) which help viruses to avoid being labeled as 'foreign' and protect them from complement-mediated neutralization and complement-enhanced antiviral adaptive immunity. In this review, we provide an overview on the structure, function and evolution of viral RCA proteins.
Collapse
Affiliation(s)
- Hina Ojha
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Hemendra Singh Panwar
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Ronald D Gorham
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Dimitrios Morikis
- Department of Bioengineering, University of California, Riverside, CA 92521, USA.
| | - Arvind Sahu
- National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
32
|
Hamed MR, Brown RJ, Zothner C, Urbanowicz RA, Mason CP, Krarup A, McClure CP, Irving WL, Ball JK, Harris M, Hickling TP, Tarr AW. Recombinant human L-ficolin directly neutralizes hepatitis C virus entry. J Innate Immun 2014; 6:676-84. [PMID: 24854201 PMCID: PMC6741592 DOI: 10.1159/000362209] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 03/16/2014] [Accepted: 03/16/2014] [Indexed: 12/25/2022] Open
Abstract
L-ficolin is a soluble pattern recognition molecule expressed by the liver that contributes to innate immune defense against microorganisms. It is well described that binding of L-ficolin to specific pathogen-associated molecular patterns activates the lectin complement pathway, resulting in opsonization and lysis of pathogens. In this study, we demonstrated that in addition to this indirect effect, L-ficolin has a direct neutralizing effect against hepatitis C virus (HCV) entry. Specific, dose-dependent binding of recombinant L-ficolin to HCV glycoproteins E1 and E2 was observed. This interaction was inhibited by soluble L-ficolin ligands. Interaction of L-ficolin with E1 and E2 potently inhibited entry of retroviral pseudoparticles bearing these glycoproteins. L-ficolin also inhibited entry of cell-cultured HCV in a calcium-dependent manner. Neutralizing concentrations of L-ficolin were found to be circulating in the serum of HCV-infected individuals. This is the first description of direct neutralization of HCV entry by a ficolin and highlights a novel role for L-ficolin as a virus entry inhibitor.
Collapse
Affiliation(s)
- Mohamed R. Hamed
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Richard J.P. Brown
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Carsten Zothner
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Richard A. Urbanowicz
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Christopher P. Mason
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Anders Krarup
- Biochemistry Department, University of Oxford, Oxford, UK
| | - C. Patrick McClure
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - William L. Irving
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Jonathan K. Ball
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Timothy P. Hickling
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Alexander W. Tarr
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| |
Collapse
|
33
|
Ball JK, Tarr AW, McKeating JA. The past, present and future of neutralizing antibodies for hepatitis C virus. Antiviral Res 2014; 105:100-11. [PMID: 24583033 PMCID: PMC4034163 DOI: 10.1016/j.antiviral.2014.02.013] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/08/2014] [Accepted: 02/13/2014] [Indexed: 12/11/2022]
Abstract
Hepatitis C virus (HCV) is a major cause of liver disease and hepatocellular carcinoma worldwide. HCV establishes a chronic infection in the majority of cases. However, some individuals clear the virus, demonstrating a protective role for the host immune response. Although new all-oral drug combinations may soon replace traditional ribavirin-interferon therapy, the emerging drug cocktails will be expensive and associated with side-effects and resistance, making a global vaccine an urgent priority. T cells are widely accepted to play an essential role in clearing acute HCV infection, whereas the role antibodies play in resolution and disease pathogenesis is less well understood. Recent studies have provided an insight into viral neutralizing determinants and the protective role of antibodies during infection. This review provides a historical perspective of the role neutralizing antibodies play in HCV infection and discusses the therapeutic benefits of antibody-based therapies. This article forms part of a symposium in Antiviral Research on "Hepatitis C: next steps toward global eradication."
Collapse
Affiliation(s)
- Jonathan K Ball
- School of Life Sciences and The Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Alexander W Tarr
- School of Life Sciences and The Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Jane A McKeating
- Viral Hepatitis Research Group and Centre for Human Virology, Institute for Biomedical Research, University of Birmingham, Birmingham B15 2TT, United Kingdom.
| |
Collapse
|
34
|
Complement and HIV-I infection/HIV-associated neurocognitive disorders. J Neurovirol 2014; 20:184-98. [PMID: 24639397 DOI: 10.1007/s13365-014-0243-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/07/2014] [Accepted: 02/11/2014] [Indexed: 10/25/2022]
Abstract
The various neurological complications associated with HIV-1 infection, specifically HIV-associated neurocognitive disorders (HAND) persist as a major public health burden worldwide. Despite the widespread use of anti-retroviral therapy, the prevalence of HAND is significantly high. HAND results from the direct effects of an HIV-1 infection as well as secondary effects of HIV-1-induced immune reaction and inflammatory response. Complement, a critical mediator of innate and acquired immunity, plays important roles in defeating many viral infections by the formation of a lytic pore or indirectly by opsonization and recruitment of phagocytes. While the role of complement in the pathogenesis of HIV-1 infection and HAND has been previously recognized for over 15 years, it has been largely underestimated thus far. Complement can be activated through HIV-1 envelope proteins, mannose-binding lectins (MBL), and anti-HIV-1 antibodies. Complement not only fights against HIV-1 infection but also enhances HIV-1 infection. In addition, HIV-1 can hijack complement regulators such as CD59 and CD55 and can utilize these regulators and factor H to escape from complement attack. Normally, complement levels in brain are much lower than plasma levels and there is no or little complement deposition in brain cells. Interestingly, local production and deposition of complement are dramatically increased in HIV-1-infected brain, indicating that complement may contribute to the pathogenesis of HAND. Here, we review the current understanding of the role of complement in HIV-1 infection and HAND, as well as potential therapeutic approaches targeting the complement system for the treatment and eradications of HIV-1 infection.
Collapse
|
35
|
HIV-1 coinfection profoundly alters intrahepatic chemokine but not inflammatory cytokine profiles in HCV-infected subjects. PLoS One 2014; 9:e86964. [PMID: 24516541 PMCID: PMC3916319 DOI: 10.1371/journal.pone.0086964] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 12/20/2013] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of accelerated liver damage in subjects coinfected with hepatitis C virus (HCV) and human immunodeficiency virus type 1 (HIV-1) remains largely unknown. Recent studies suggest that ongoing chronic liver inflammation is responsible for the liver injury in HCV-infected patients. We aimed to determine whether HIV-1 coinfection altered intrahepatic inflammatory profiles in HCV infection, thereby hastening liver damage. We used a real-time RT-PCR-based array to comparatively analyze intrahepatic inflammation gene profiles in liver biopsy specimens from HCV-infected (n = 16), HCV/HIV-1-coinfected (n = 8) and uninfected (n = 8) individuals. We then used human hepatocytes to study the molecular mechanisms underlying alternations of the inflammatory profiles. Compared with uninfected individuals, HCV infection and HCV/HIV-1 coinfection markedly altered expression of 59.5% and 50.0% of 84 inflammation-related genes tested, respectively. Among these genes affected, HCV infection up-regulated the expression of 24 genes and down-regulated the expression of 26 genes, whereas HCV/HIV-1 coinfection up-regulated the expression of 21 genes and down-regulated the expression of 21 genes. Compared with HCV infection, HCV/HIV-1 coinfection did not dramatically affect intrahepatic gene expression profiles of cytokines and their receptors, but profoundly altered expression of several chemokine genes including up-regulation of the CXCR3-associated chemokines. Human hepatocytes produced these chemokines in response to virus-related microbial translocation, viral protein stimulation, and antiviral immune responses. Conclusions HIV-1 coinfection profoundly alters intrahepatic chemokine but not cytokine profiles in HCV-infected subjects. The altered chemokines may orchestrate the tissue-specific and cell-selective trafficking of immune cells and autoimmunity to accelerate liver disease in HCV/HIV-1 coinfection.
Collapse
|
36
|
Mawatari S, Uto H, Ido A, Nakashima K, Suzuki T, Kanmura S, Kumagai K, Oda K, Tabu K, Tamai T, Moriuchi A, Oketani M, Shimada Y, Sudoh M, Shoji I, Tsubouchi H. Hepatitis C virus NS3/4A protease inhibits complement activation by cleaving complement component 4. PLoS One 2013; 8:e82094. [PMID: 24349192 PMCID: PMC3861371 DOI: 10.1371/journal.pone.0082094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/11/2013] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND It has been hypothesized that persistent hepatitis C virus (HCV) infection is mediated in part by viral proteins that abrogate the host immune response, including the complement system, but the precise mechanisms are not well understood. We investigated whether HCV proteins are involved in the fragmentation of complement component 4 (C4), composed of subunits C4α, C4β, and C4γ, and the role of HCV proteins in complement activation. METHODS Human C4 was incubated with HCV nonstructural (NS) 3/4A protease, core, or NS5. Samples were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then subjected to peptide sequencing. The activity of the classical complement pathway was examined using an erythrocyte hemolysis assay. The cleavage pattern of C4 in NS3/4A-expressing and HCV-infected cells, respectively, was also examined. RESULTS HCV NS3/4A protease cleaved C4γ in a concentration-dependent manner, but viral core and NS5 did not. A specific inhibitor of NS3/4A protease reduced C4γ cleavage. NS3/4A protease-mediated cleavage of C4 inhibited classical pathway activation, which was abrogated by a NS3/4A protease inhibitor. In addition, co-transfection of cells with C4 and wild-type NS3/4A, but not a catalytic-site mutant of NS3/4A, produced cleaved C4γ fragments. Such C4 processing, with a concomitant reduction in levels of full-length C4γ, was also observed in HCV-infected cells expressing C4. CONCLUSIONS C4 is a novel cellular substrate of the HCV NS3/4A protease. Understanding disturbances in the complement system mediated by NS3/4A protease may provide new insights into the mechanisms underlying persistent HCV infection.
Collapse
Affiliation(s)
- Seiichi Mawatari
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Hirofumi Uto
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
- * E-mail:
| | - Akio Ido
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Kenji Nakashima
- Department of Infectious Diseases, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Tetsuro Suzuki
- Department of Infectious Diseases, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Shuji Kanmura
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Kotaro Kumagai
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Kohei Oda
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Kazuaki Tabu
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Tsutomu Tamai
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Akihiro Moriuchi
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Makoto Oketani
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima, Japan
| | - Yuko Shimada
- Miyazaki Prefectural Industrial Support Foundation, Miyazaki, Miyazaki, Japan
| | - Masayuki Sudoh
- Kamakura Research Division, Chugai Pharmaceutical, Co. Ltd., Kamakura, Kanagawa, Japan
| | - Ikuo Shoji
- Division of Microbiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hirohito Tsubouchi
- Department of HGF Tissue Repair and Regenerative Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
37
|
Du Y, Teng X, Wang N, Zhang X, Chen J, Ding P, Qiao Q, Wang Q, Zhang L, Yang C, Yang Z, Chu Y, Du X, Zhou X, Hu W. NF-κB and enhancer-binding CREB protein scaffolded by CREB-binding protein (CBP)/p300 proteins regulate CD59 protein expression to protect cells from complement attack. J Biol Chem 2013; 289:2711-24. [PMID: 24338025 DOI: 10.1074/jbc.m113.525501] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The complement system can be activated spontaneously for immune surveillance or induced to clear invading pathogens, in which the membrane attack complex (MAC, C5b-9) plays a critical role. CD59 is the sole membrane complement regulatory protein (mCRP) that restricts MAC assembly. CD59, therefore, protects innocent host cells from attacks by the complement system, and host cells require the constitutive and inducible expression of CD59 to protect themselves from deleterious destruction by complement. However, the mechanisms that underlie CD59 regulation remain largely unknown. In this study we demonstrate that the widely expressed transcription factor Sp1 may regulate the constitutive expression of CD59, whereas CREB-binding protein (CBP)/p300 bridge NF-κB and CREB, which surprisingly functions as an enhancer-binding protein to induce the up-regulation of CD59 during in lipopolysaccharide (LPS)-triggered complement activation, thus conferring host defense against further MAC-mediated destruction. Moreover, individual treatment with LPS, TNF-α, and the complement activation products (sublytic MAC (SC5b-9) and C5a) could increase the expression of CD59 mainly by activating NF-κB and CREB signaling pathways. Together, our findings identify a novel gene regulation mechanism involving CBP/p300, NF-κB, and CREB; this mechanism suggests potential drug targets for controlling various complement-related human diseases.
Collapse
Affiliation(s)
- Yiqun Du
- From the Cancer Institute, Collaborative Innovation Center of Cancer Medicine, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Douradinha B, McBurney SP, Soares de Melo KM, Smith AP, Krishna NK, Barratt-Boyes SM, Evans JD, Nascimento EJM, Marques ETA. C1q binding to dengue virus decreases levels of infection and inflammatory molecules transcription in THP-1 cells. Virus Res 2013; 179:231-4. [PMID: 24246304 DOI: 10.1016/j.virusres.2013.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/03/2013] [Accepted: 11/07/2013] [Indexed: 11/28/2022]
Abstract
Dengue virus infection elicits a spectrum of clinical presentations ranging from asymptomatic to severe disease. The mechanisms leading to severe dengue are not known, however it has been reported that the complement system is hyper-activated in severe dengue. Screening of complement proteins demonstrated that C1q, a pattern recognition molecule, can bind directly to dengue virus envelope protein and to whole dengue virus serotype 2. Incubation of dengue virus serotype 2 with C1q prior to infection of THP-1 cells led to decreased virus infectivity and modulation of mRNA expression of immunoregulatory molecules suggesting reduced inflammatory responses.
Collapse
Affiliation(s)
- Bruno Douradinha
- Fondazione Ri.MED, Via Bandiera 11, 90133 Palermo, Italy; University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA.
| | - Sean P McBurney
- University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA
| | - Klecia M Soares de Melo
- University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA; Virology and Experimental Therapy Laboratory, Aggeu Magalhães Research Center, Fiocruz, Recife, Pernambuco 50670-420, Brazil
| | - Amanda P Smith
- University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA
| | - Neel K Krishna
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Lewis Hall, Room 3184, 700W. Olney Road, Norfolk, VA 23507, USA
| | - Simon M Barratt-Boyes
- University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA; Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15261, USA
| | - Jared D Evans
- University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 523 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219, USA
| | - Eduardo J M Nascimento
- University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA; Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15261, USA
| | - Ernesto T A Marques
- University of Pittsburgh Center for Vaccine Research, 3501 Fifth Avenue, 9th floor, Pittsburgh, PA 15261, USA; Virology and Experimental Therapy Laboratory, Aggeu Magalhães Research Center, Fiocruz, Recife, Pernambuco 50670-420, Brazil; Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15261, USA
| |
Collapse
|
39
|
Allison AB, Mead DG, Palacios GF, Tesh RB, Holmes EC. Gene duplication and phylogeography of North American members of the Hart Park serogroup of avian rhabdoviruses. Virology 2013; 448:284-92. [PMID: 24314659 PMCID: PMC3873333 DOI: 10.1016/j.virol.2013.10.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Revised: 06/11/2013] [Accepted: 10/17/2013] [Indexed: 12/28/2022]
Abstract
Flanders virus (FLAV) and Hart Park virus (HPV) are rhabdoviruses that circulate in mosquito–bird cycles in the eastern and western United States, respectively, and constitute the only two North American representatives of the Hart Park serogroup. Previously, it was suggested that FLAV is unique among the rhabdoviruses in that it contains two pseudogenes located between the P and M genes, while the cognate sequence for HPV has been lacking. Herein, we demonstrate that FLAV and HPV do not contain pseudogenes in this region, but encode three small functional proteins designated as U1–U3 that apparently arose by gene duplication. To further investigate the U1–U3 region, we conducted the first large-scale evolutionary analysis of a member of the Hart Park serogroup by analyzing over 100 spatially and temporally distinct FLAV isolates. Our phylogeographic analysis demonstrates that although FLAV appears to be slowly evolving, phylogenetically divergent lineages co-circulate sympatrically. Flanders virus (FLAV) does not contain pseudogenes as previously reported. The FLAV U1–U3 proteins arose by gene duplication. The SH protein of FLAV is tentatively expressed by coupled translation. Distinct lineages of FLAV circulate sympatrically in the United States. Histone H4 and cyclophilin A are apparently incorporated into FLAV particles.
Collapse
Affiliation(s)
- Andrew B Allison
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA; Southeastern Cooperative Wildlife Disease Study, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.
| | | | | | | | | |
Collapse
|
40
|
Hepatitis C virus infection upregulates CD55 expression on the hepatocyte surface and promotes association with virus particles. J Virol 2013; 87:7902-10. [PMID: 23658447 DOI: 10.1128/jvi.00917-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
CD55 limits excessive complement activation on the host cell surface by accelerating the decay of C3 convertases. In this study, we observed that hepatitis C virus (HCV) infection of hepatocytes or HCV core protein expression in transfected hepatocytes upregulated CD55 expression at the mRNA and protein levels. Further analysis suggested that the HCV core protein or full-length (FL) genome enhanced CD55 promoter activity in a luciferase-based assay, which was further augmented in the presence of interleukin-6. Mutation of the CREB or SP-1 binding site on the CD55 promoter impaired HCV core protein-mediated upregulation of CD55. HCV-infected or core protein-transfected Huh7.5 cells displayed greater viability in the presence of CD81 and CD55 antibodies and complement. Biochemical analysis revealed that CD55 was associated with cell culture-grown HCV after purification by sucrose density gradient ultracentrifugation. Consistent with this, a polyclonal antibody to CD55 captured cell culture-grown HCV. Blocking antibodies against CD55 or virus envelope glycoproteins in the presence of normal human serum as a source of complement inhibited HCV infection. The inhibition was enhanced in the presence of both the antibodies and serum complement. Collectively, these results suggest that HCV induces and associates with a negative regulator of the complement pathway, a likely mechanism for immune evasion.
Collapse
|
41
|
Hepatitis C virus suppresses C9 complement synthesis and impairs membrane attack complex function. J Virol 2013; 87:5858-67. [PMID: 23487461 DOI: 10.1128/jvi.00174-13] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatitis C virus (HCV) proteins inhibit complement component expression, which may attenuate immunity against infection. In this study, we examined whether HCV regulates the membrane attack complex (MAC) via complement component C9. MAC is composed of C5b to C9 (C5b-9) and mediates cell lysis of invaded pathogens. Liver biopsy specimens from chronically HCV-infected patients exhibited a lower level of C9 mRNA expression than liver biopsy specimens from unrelated disease or healthy control human liver RNA. Hepatocytes infected with cell culture-grown HCV or expressing HCV core protein also displayed significant repression of C9 mRNA and protein levels. Promoter analysis suggested that the T cell factor-4 (TCF-4E) transcription factor is responsible for HCV core-mediated C9 promoter regulation. Sera from chronically HCV-infected patients displayed a lower level of C5b-9 and a reduced antimicrobial effect on model organisms compared to unrelated patient sera or sera from healthy volunteers. Together, these results for C9 regulation by HCV core protein coupled with functional impairment of the membrane attack complex underscore HCV-mediated attenuation of immune mechanisms.
Collapse
|
42
|
Shuang F, Luo Y, Xiong XP, Weng S, Li Y, He J, Dong C. Virions proteins of an RSIV-type megalocytivirus from spotted knifejaw Oplegnathus punctatus (SKIV-ZJ07). Virology 2013; 437:89-99. [DOI: 10.1016/j.virol.2012.12.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/24/2012] [Accepted: 12/29/2012] [Indexed: 11/29/2022]
|
43
|
Ejaz A, Steinmann E, Bánki Z, Anggakusuma, Khalid S, Lengauer S, Wilhelm C, Zoller H, Schloegl A, Steinmann J, Grabski E, Kleines M, Pietschmann T, Stoiber H. Specific acquisition of functional CD59 but not CD46 or CD55 by hepatitis C virus. PLoS One 2012; 7:e45770. [PMID: 23049856 PMCID: PMC3458075 DOI: 10.1371/journal.pone.0045770] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 08/22/2012] [Indexed: 01/07/2023] Open
Abstract
Viruses of different families encode for regulators of the complement system (RCAs) or acquire such RCAs from the host to get protection against complement-mediated lysis (CML). As hepatitis C virus (HCV) shares no genetic similarity to any known RCA and is detectable at high titers in sera of infected individuals, we investigated whether HCV has adapted host-derived RCAs to resist CML. Here we report that HCV selectively incorporates CD59 while neither CD55, nor CD46 are associated with the virus. The presence of CD59 was shown by capture assays using patient- and cell culture-derived HCV isolates. Association of CD59 with HCV was further confirmed by Western blot analysis using purified viral supernatants from infected Huh 7.5 cells. HCV captured by antibodies specific for CD59 remained infectious for Huh 7.5 cells. In addition, blocking of CD59 in the presence of active complement reduced the titer of HCV most likely due to CML. HCV produced in CD59 knock-down cells were more significantly susceptible to CML compared to wild type virus, but neither replication, assembly nor infectivity of the virus seemed to be impaired in the absence of CD59. In summary our data indicate that HCV incorporates selectively CD59 in its envelope to gain resistance to CML in serum of infected individuals.
Collapse
Affiliation(s)
- Asim Ejaz
- Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tissue-specific transplantation antigen P35B (TSTA3) immune response-mediated metabolism coupling cell cycle to postreplication repair network in no-tumor hepatitis/cirrhotic tissues (HBV or HCV infection) by biocomputation. Immunol Res 2012; 52:258-68. [PMID: 22528125 DOI: 10.1007/s12026-012-8337-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We constructed the low-expression tissue-specific transplantation antigen P35B (TSTA3) immune response-mediated metabolism coupling cell cycle to postreplication repair network in no-tumor hepatitis/cirrhotic tissues (HBV or HCV infection) compared with high-expression (fold change ≥ 2) human hepatocellular carcinoma in GEO data set, by using integration of gene regulatory network inference method with gene ontology analysis of TSTA3-activated up- and downstream networks. Our results showed TSTA3 upstream-activated CCNB2, CKS1B, ELAVL3, GAS7, NQO1, NTN1, OCRL, PLA2G1B, REG3A, SSTR5, etc. and TSTA3 downstream-activated BAP1, BRCA1, CCL20, MCM2, MS4A2, NTN1, REG1A, TP53I11, VCAN, SLC16A3, etc. in no-tumor hepatitis/cirrhotic tissues. TSTA3-activated network enhanced the regulation of apoptosis, cyclin-dependent protein kinase activity, cell migration, insulin secretion, transcription, cell division, cell proliferation, DNA replication, postreplication repair, cell differentiation, T-cell homeostasis, neutrophil-mediated immunity, neutrophil chemotaxis, interleukin-8 production, inflammatory response, immune response, B-cell activation, humoral immune response, actin filament organization, xenobiotic metabolism, lipid metabolism, phospholipid metabolism, leukotriene biosynthesis, organismal lipid catabolism, phosphatidylcholine metabolism, arachidonic acid secretion, activation of phospholipase A2, deoxyribonucleotide biosynthesis, heterophilic cell adhesion, activation of MAPK activity, signal transduction by p53 class mediator resulting in transcription of p21 class mediator, G-protein-coupled receptor protein signaling pathway, response to toxin, acute-phase response, DNA damage response, intercellular junction assembly, cell communication, and cell recognition, as a result of inducing immune response-mediated metabolism coupling cell cycle to postreplication repair in no-tumor hepatitis/cirrhotic tissues.
Collapse
|
45
|
Tarr AW, Urbanowicz RA, Ball JK. The role of humoral innate immunity in hepatitis C virus infection. Viruses 2012; 4:1-27. [PMID: 22355450 PMCID: PMC3280516 DOI: 10.3390/v4010001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 12/21/2011] [Accepted: 12/23/2011] [Indexed: 12/19/2022] Open
Abstract
Infection with Hepatitis C Virus (HCV) causes chronic disease in approximately 80% of cases, resulting in chronic inflammation and cirrhosis. Current treatments are not completely effective, and a vaccine has yet to be developed. Spontaneous resolution of infection is associated with effective host adaptive immunity to HCV, including production of both HCV-specific T cells and neutralizing antibodies. However, the supporting role of soluble innate factors in protection against HCV is less well understood. The innate immune system provides an immediate line of defense against infections, triggering inflammation and playing a critical role in activating adaptive immunity. Innate immunity comprises both cellular and humoral components, the humoral arm consisting of pattern recognition molecules such as complement C1q, collectins and ficolins. These molecules activate the complement cascade, neutralize pathogens, and recruit antigen presenting cells. Here we review the current understanding of anti-viral components of the humoral innate immune system that play a similar role to antibodies, describing their role in immunity to HCV and their potential contribution to HCV pathogenesis.
Collapse
Affiliation(s)
- Alexander W. Tarr
- Biomedical Research Unit in Gastroenterology, School of Molecular Medical Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; (R.A.U.); (J.K.B.)
| | | | | |
Collapse
|