1
|
Ye S, Liang Y, Chang Y, Lai B, Zhong J. Dengue Virus Replicative-Form dsRNA Is Recognized by Both RIG-I and MDA5 to Activate Innate Immunity. J Med Virol 2025; 97:e70194. [PMID: 39873327 DOI: 10.1002/jmv.70194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
RIG-I like receptors (RLRs) are a family of cytosolic RNA sensors that sense RNA virus infection to activate innate immune response. It is generally believed that different RNA viruses are recognized by either RIG-I or MDA5, two important RLR members, depending on the nature of pathogen-associated molecular patterns (PAMPs) that are generated by RNA virus replication. Dengue virus (DENV) is an important RNA virus causing serious human diseases. Despite extensive investigations, the molecular basis of the DENV PAMP recognized by the host RLR has been poorly defined. Here, we demonstrated that the DENV infection-induced interferon response is dependent upon both RIG-I and MDA5, with RIG-I playing a predominant role. Next we purified the DENV PAMP RNA from the DENV-infected cells, and demonstrated that the purified DENV PAMP is viral full-length double-stranded RNA bearing 5'ppp modifications, likely representing the viral replicative-form RNA. Finally, we confirmed the nature of the DENV PAMP by reconstituting the viral replicative-form RNA from in vitro synthesized DENV genomic RNA. In conclusion, our work not only defined the molecular basis of the RLR-PAMP interaction during DENV infection, but also revealed the previously underappreciated recognition of a distinct moiety of the same PAMP by different RLRs in innate immunity against RNA viruses.
Collapse
Affiliation(s)
- Sichao Ye
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yisha Liang
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yu Chang
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bailiang Lai
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Jin Zhong
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
2
|
Coelho SVA, Augusto FM, de Arruda LB. Potential Pathways and Pathophysiological Implications of Viral Infection-Driven Activation of Kallikrein-Kinin System (KKS). Viruses 2024; 16:245. [PMID: 38400022 PMCID: PMC10892958 DOI: 10.3390/v16020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Microcirculatory and coagulation disturbances commonly occur as pathological manifestations of systemic viral infections. Research exploring the role of the kallikrein-kinin system (KKS) in flavivirus infections has recently linked microvascular dysfunctions to bradykinin (BK)-induced signaling of B2R, a G protein-coupled receptor (GPCR) constitutively expressed by endothelial cells. The relevance of KKS activation as an innate response to viral infections has gained increasing attention, particularly after the reports regarding thrombogenic events during COVID-19. BK receptor (B2R and B1R) signal transduction results in vascular permeability, edema formation, angiogenesis, and pain. Recent findings unveiling the role of KKS in viral pathogenesis include evidence of increased activation of KKS with elevated levels of BK and its metabolites in both intravascular and tissue milieu, as well as reports demonstrating that virus replication stimulates BKR expression. In this review, we will discuss the mechanisms triggered by virus replication and by virus-induced inflammatory responses that may stimulate KKS. We also explore how KKS activation and BK signaling may impact virus pathogenesis and further discuss the potential therapeutic application of BKR antagonists in the treatment of hemorrhagic and respiratory diseases.
Collapse
Affiliation(s)
- Sharton Vinícius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | | | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
3
|
Blackhurst BM, Funk KE. Molecular and Cellular Mechanisms Underlying Neurologic Manifestations of Mosquito-Borne Flavivirus Infections. Viruses 2023; 15:2200. [PMID: 38005878 PMCID: PMC10674799 DOI: 10.3390/v15112200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Flaviviruses are a family of enveloped viruses with a positive-sense RNA genome, transmitted by arthropod vectors. These viruses are known for their broad cellular tropism leading to infection of multiple body systems, which can include the central nervous system. Neurologic effects of flavivirus infection can arise during both acute and post-acute infectious periods; however, the molecular and cellular mechanisms underlying post-acute sequelae are not fully understood. Here, we review recent studies that have examined molecular and cellular mechanisms that may contribute to neurologic sequelae following infection with the West Nile virus, Japanese encephalitis virus, Zika virus, dengue virus, and St. Louis encephalitis virus. Neuronal death, either from direct infection or due to the resultant inflammatory response, is a common mechanism by which flavivirus infection can lead to neurologic impairment. Other types of cellular damage, such as oxidative stress and DNA damage, appear to be more specific to certain viruses. This article aims to highlight mechanisms of cellular damage that are common across several flavivirus members and mechanisms that are more unique to specific members. Our goal is to inspire further research to improve understanding of this area in the hope of identifying treatment options for flavivirus-associated neurologic changes.
Collapse
Affiliation(s)
| | - Kristen E. Funk
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
4
|
Jaeger AS, Marano J, Riemersma KK, Castaneda D, Pritchard EM, Pritchard JC, Bohm EK, Baczenas JJ, O'Connor SL, Weger-Lucarelli J, Friedrich TC, Aliota MT. Gain without pain: adaptation and increased virulence of Zika virus in vertebrate host without fitness cost in mosquito vector. J Virol 2023; 97:e0116223. [PMID: 37800949 PMCID: PMC10653995 DOI: 10.1128/jvi.01162-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
IMPORTANCE Previously, we modeled direct transmission chains of Zika virus (ZIKV) by serially passaging ZIKV in mice and mosquitoes and found that direct mouse transmission chains selected for viruses with increased virulence in mice and the acquisition of non-synonymous amino acid substitutions. Here, we show that these same mouse-passaged viruses also maintain fitness and transmission capacity in mosquitoes. We used infectious clone-derived viruses to demonstrate that the substitution in nonstructural protein 4A contributes to increased virulence in mice.
Collapse
Affiliation(s)
- Anna S. Jaeger
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Jeffrey Marano
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Kasen K. Riemersma
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David Castaneda
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Elise M. Pritchard
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Julia C. Pritchard
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Ellie K. Bohm
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - John J. Baczenas
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Shelby L. O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| |
Collapse
|
5
|
Modak A, Mishra SR, Awasthi M, Sreedevi S, Sobha A, Aravind A, Kuppusamy K, Sreekumar E. Higher-temperature-adapted dengue virus serotype 2 strain exhibits enhanced virulence in AG129 mouse model. FASEB J 2023; 37:e23062. [PMID: 37389962 DOI: 10.1096/fj.202300098r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/13/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
The factors that drive dengue virus (DENV) evolution, and selection of virulent variants are yet not clear. Higher environmental temperature shortens DENV extrinsic incubation period in mosquitoes, increases human transmission, and plays a critical role in outbreak dynamics. In the present study, we looked at the effect of temperature in altering the virus virulence. We found that DENV cultured at a higher temperature in C6/36 mosquito cells was significantly more virulent than the virus grown at a lower temperature. In a mouse model, the virulent strain induced enhanced viremia and aggressive disease with a short course, hemorrhage, severe vascular permeability, and death. Higher inflammatory cytokine response, thrombocytopenia, and severe histopathological changes in vital organs such as heart, liver, and kidney were hallmarks of the disease. Importantly, it required only a few passages for the virus to acquire a quasi-species population harboring virulence-imparting mutations. Whole genome comparison with a lower temperature passaged strain identified key genomic changes in the structural protein-coding regions as well as in the 3'UTR of the viral genome. Our results point out that virulence-enhancing genetic changes could occur in the dengue virus genome under enhanced growth temperature conditions in mosquito cells.
Collapse
Affiliation(s)
- Ayan Modak
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Srishti Rajkumar Mishra
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Mansi Awasthi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Sreeja Sreedevi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Archana Sobha
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Arya Aravind
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Krithiga Kuppusamy
- Bioscience Research & Training Centre (BRTC), Kerala Veterinary and Animal Sciences University, Bio360 Life Sciences Park, Thiruvananthapuram, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology (IAV), Bio360 Life Sciences Park, Thiruvananthapuram, India
| |
Collapse
|
6
|
Baris A, Fraile-Bethencourt E, Eubanks J, Khou S, Anand S. Thymidine phosphorylase facilitates retinoic acid inducible gene-I induced endothelial dysfunction. Cell Death Dis 2023; 14:294. [PMID: 37100811 PMCID: PMC10131517 DOI: 10.1038/s41419-023-05821-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023]
Abstract
Activation of nucleic acid sensors in endothelial cells (ECs) has been shown to drive inflammation across pathologies including cancer, atherosclerosis and obesity. We previously showed that enhancing cytosolic DNA sensing by inhibiting three prime exonuclease 1 (TREX1) in ECs led to EC dysfunction and impaired angiogenesis. Here we show that activation of a cytosolic RNA sensor, Retinoic acid Induced Gene 1 (RIG-I) diminishes EC survival, angiogenesis and triggers tissue specific gene expression programs. We discovered a RIG-I dependent 7 gene signature that affects angiogenesis, inflammation and coagulation. Among these, we identified the thymidine phosphorylase TYMP as a key mediator of RIG-I induced EC dysfunction via its regulation of a subset of interferon stimulated genes. Our RIG-I induced gene signature was also conserved in the context of human diseases - in lung cancer vasculature and herpesvirus infection of lung endothelial cells. Pharmacological or genetic inhibition of TYMP rescues RIG-I induced EC death, migration arrest and restores sprouting angiogenesis. Interestingly, using RNAseq we identified a gene expression program that was RIG-I induced but TYMP dependent. Analysis of this dataset indicated that IRF1 and IRF8 dependent transcription is diminished in RIG-I activated cells when TYMP is inhibited. Functional RNAi screen of our TYMP dependent EC genes, we found that a group of 5 genes - Flot1, Ccl5, Vars2, Samd9l and Ube2l6 are critical for endothelial cell death mediated by RIG-I activation. Our observations identify mechanisms by which RIG-I drives EC dysfunction and define pathways that can be pharmacologically targeted to ameliorate RIG-I induced vascular inflammation.
Collapse
Affiliation(s)
- Adrian Baris
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Eugenia Fraile-Bethencourt
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Jaiden Eubanks
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Sokchea Khou
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Sudarshan Anand
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA.
- Department of Radiation Medicine, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA.
| |
Collapse
|
7
|
Jaeger AS, Marano J, Riemersma K, Castañeda D, Pritchard E, Pritchard J, Bohm EK, Baczenas JJ, O’Connor SL, Weger-Lucarelli J, Friedrich TC, Aliota MT. Gain without pain: Adaptation and increased virulence of Zika virus in vertebrate host without fitness cost in mosquito vector. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.20.533515. [PMID: 36993525 PMCID: PMC10055270 DOI: 10.1101/2023.03.20.533515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Zika virus (ZIKV) is now in a post-pandemic period, for which the potential for re-emergence and future spread is unknown. Adding to this uncertainty is the unique capacity of ZIKV to directly transmit between humans via sexual transmission. Recently, we demonstrated that direct transmission of ZIKV between vertebrate hosts leads to rapid adaptation resulting in enhanced virulence in mice and the emergence of three amino acid substitutions (NS2A-A117V, NS2A-A117T, and NS4A-E19G) shared among all vertebrate-passaged lineages. Here, we further characterized these host-adapted viruses and found that vertebrate-passaged viruses also have enhanced transmission potential in mosquitoes. To understand the contribution of genetic changes to the enhanced virulence and transmission phenotype, we engineered these amino acid substitutions, singly and in combination, into a ZIKV infectious clone. We found that NS4A-E19G contributed to the enhanced virulence and mortality phenotype in mice. Further analyses revealed that NS4A-E19G results in increased neurotropism and distinct innate immune signaling patterns in the brain. None of the substitutions contributed to changes in transmission potential in mosquitoes. Together, these findings suggest that direct transmission chains could enable the emergence of more virulent ZIKV strains without compromising mosquito transmission capacity, although the underlying genetics of these adaptations are complex.
Collapse
Affiliation(s)
- Anna S. Jaeger
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Jeffrey Marano
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University
| | - Kasen Riemersma
- Department of Pathobiological Sciences, University of Wisconsin-Madison
| | - David Castañeda
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Elise Pritchard
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Julia Pritchard
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Ellie K. Bohm
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - John J. Baczenas
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison
- Wisconsin National Primate Research Center, University of Wisconsin-Madison
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| |
Collapse
|
8
|
Atalis A, Keenum MC, Pandey B, Beach A, Pradhan P, Vantucci C, O'Farrell L, Noel R, Jain R, Hosten J, Smith C, Kramer L, Jimenez A, Ochoa MA, Frey D, Roy K. Nanoparticle-delivered TLR4 and RIG-I agonists enhance immune response to SARS-CoV-2 subunit vaccine. J Control Release 2022; 347:476-488. [PMID: 35577151 PMCID: PMC9121740 DOI: 10.1016/j.jconrel.2022.05.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/04/2022] [Accepted: 05/10/2022] [Indexed: 01/25/2023]
Abstract
Despite success in vaccinating populations against SARS-CoV-2, concerns about immunity duration, continued efficacy against emerging variants, protection from infection and transmission, and worldwide vaccine availability remain. Molecular adjuvants targeting pattern recognition receptors (PRRs) on antigen-presenting cells (APCs) could improve and broaden the efficacy and durability of vaccine responses. Native SARS-CoV-2 infection stimulates various PRRs, including toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors. We hypothesized that targeting PRRs using molecular adjuvants on nanoparticles (NPs) along with a stabilized spike protein antigen could stimulate broad and efficient immune responses. Adjuvants targeting TLR4 (MPLA), TLR7/8 (R848), TLR9 (CpG), and RIG-I (PUUC) delivered on degradable polymer NPs were combined with the S1 subunit of spike protein and assessed in vitro with isogeneic mixed lymphocyte reactions (isoMLRs). For in vivo studies, the adjuvant-NPs were combined with stabilized spike protein or spike-conjugated NPs and assessed using a two-dose intranasal or intramuscular vaccination model in mice. Combination adjuvant-NPs simultaneously targeting TLR and RIG-I receptors (MPLA+PUUC, CpG+PUUC, and R848+PUUC) differentially induced T cell proliferation and increased proinflammatory cytokine secretion by APCs in vitro. When delivered intranasally, MPLA+PUUC NPs enhanced CD4+CD44+ activated memory T cell responses against spike protein in the lungs while MPLA NPs increased anti-spike IgA in the bronchoalveolar (BAL) fluid and IgG in the blood. Following intramuscular delivery, PUUC NPs induced strong humoral immune responses, characterized by increases in anti-spike IgG in the blood and germinal center B cell populations (GL7+ and BCL6+ B cells) in the draining lymph nodes (dLNs). MPLA+PUUC NPs further boosted spike protein-neutralizing antibody titers and T follicular helper cell populations in the dLNs. These results suggest that protein subunit vaccines with particle-delivered molecular adjuvants targeting TLR4 and RIG-I could lead to robust and unique route-specific adaptive immune responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Alexandra Atalis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mark C Keenum
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Bhawana Pandey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Alexander Beach
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Pallab Pradhan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Casey Vantucci
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Laura O'Farrell
- Physiological Research Laboratory, Georgia Institute of Technology, Atlanta, GA, USA
| | - Richard Noel
- Physiological Research Laboratory, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ritika Jain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Justin Hosten
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Clinton Smith
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Liana Kramer
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Angela Jimenez
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Miguel Armenta Ochoa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - David Frey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
9
|
Wang H, Yin J, Gu X, Shao W, Jia Z, Chen H, Xia W. Immune Regulator Retinoic Acid-Inducible Gene I (RIG-I) in the Pathogenesis of Cardiovascular Disease. Front Immunol 2022; 13:893204. [PMID: 35693778 PMCID: PMC9178270 DOI: 10.3389/fimmu.2022.893204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Retinoic acid-inducible gene I (RIG-I) is a cytosolic pattern recognition receptor that contains two CARD domains, an RNA helicase domain, and a C-terminal domain. RIG-I initiates antiviral innate immunity by recognizing exogenous viral RNAs/DNAs. However, some studies have reported that RIG-I activation leads to damage in various organs and tissues in diverse circumstances. Recent studies have shown that RIG-I is involved in cancer, lupus nephritis, immunoglobulin A nephropathy, Crohn's disease, and atherosclerosis. These reports indicate that RIG-I not only participates in antiviral signaling pathways but also exerts an influence on non-viral infectious diseases. RIG-I is widely expressed in immune and non-immune cells including smooth muscle cells, endothelial cells, and cardiomyocytes. A succinct overview of RIG-I and its signaling pathways, with respect to the cardiovascular system, will aid in the development of novel therapeutics for cardiovascular diseases. In this review, we summarize the structure, activation, signaling pathways, and role of RIG-I in cardiovascular diseases.
Collapse
Affiliation(s)
- Hao Wang
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Yin
- Department of Cardiology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyan Gu
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Wenhui Shao
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Hongbing Chen
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Xia
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Bhide K, Mochnáčová E, Tkáčová Z, Petroušková P, Kulkarni A, Bhide M. Signaling events evoked by domain III of envelop glycoprotein of tick-borne encephalitis virus and West Nile virus in human brain microvascular endothelial cells. Sci Rep 2022; 12:8863. [PMID: 35614140 PMCID: PMC9133079 DOI: 10.1038/s41598-022-13043-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022] Open
Abstract
Tick-borne encephalitis virus and West Nile virus can cross the blood–brain barrier via hematogenous route. The attachment of a virion to the cells of a neurovascular unit, which is mediated by domain III of glycoprotein E, initiates a series of events that may aid viral entry. Thus, we sought to uncover the post-attachment biological events elicited in brain microvascular endothelial cells by domain III. RNA sequencing of cells treated with DIII of TBEV and WNV showed significant alteration in the expression of 309 and 1076 genes, respectively. Pathway analysis revealed activation of the TAM receptor pathway. Several genes that regulate tight-junction integrity were also activated, including pro-inflammatory cytokines and chemokines, cell-adhesion molecules, claudins, and matrix metalloprotease (mainly ADAM17). Results also indicate activation of a pro-apoptotic pathway. TLR2 was upregulated in both cases, but MyD88 was not. In the case of TBEV DIII, a MyD88 independent pathway was activated. Furthermore, both cases showed dramatic dysregulation of IFN and IFN-induced genes. Results strongly suggest that the virus contact to the cell surface emanates a series of events namely viral attachment and diffusion, breakdown of tight junctions, induction of virus uptake, apoptosis, reorganization of the extracellular-matrix, and activation of the innate immune system.
Collapse
Affiliation(s)
- Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Evelína Mochnáčová
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Zuzana Tkáčová
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Patrícia Petroušková
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic
| | - Amod Kulkarni
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic.,Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Kosice, Slovak Republic. .,Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
11
|
Meuren LM, Prestes EB, Papa MP, de Carvalho LRP, Mustafá YM, da Costa LS, Da Poian AT, Bozza MT, Arruda LB. Infection of Endothelial Cells by Dengue Virus Induces ROS Production by Different Sources Affecting Virus Replication, Cellular Activation, Death and Vascular Permeability. Front Immunol 2022; 13:810376. [PMID: 35185902 PMCID: PMC8847576 DOI: 10.3389/fimmu.2022.810376] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/11/2022] [Indexed: 01/20/2023] Open
Abstract
Exacerbated inflammatory response and altered vascular function are hallmarks of dengue disease. Reactive oxygen species (ROS) production has been associated to endothelial barrier disturbance and microvascular alteration in distinct pathological conditions. Increased ROS has been reported in in vitro models of dengue virus (DENV) infection, but its impact for endothelial cell physiology had not been fully investigated. Our group had previously demonstrated that infection of human brain microvascular endothelial cells (HBMEC) with DENV results in the activation of RNA sensors and production of proinflammatory cytokines, which culminate in cell death and endothelial permeability. Here, we evaluated the role of mitochondrial function and NADPH oxidase (NOX) activation for ROS generation in HBMEC infected by DENV and investigated whether altered cellular physiology could be a consequence of virus-induced oxidative stress. DENV-infected HBMECs showed a decrease in the maximal respiratory capacity and altered membrane potential, indicating functional mitochondrial alteration, what might be related to mtROS production. Indeed, mtROS was detected at later time points after infection. Specific inhibition of mtROS diminished virus replication, cell death, and endothelial permeability, but did not affect cytokine production. On the other hand, inhibition of NOX-associated ROS production decreased virus replication and cell death, as well as the secretion of inflammatory cytokines, including IL-6, IL-8, and CCL5. These results demonstrated that DENV replication in endothelial cells induces ROS production by different pathways, which impacts biological functions that might be relevant for dengue pathogenesis. Those data also indicate oxidative stress events as relevant therapeutical targets to avoid vascular permeability, inflammation, and neuroinvasion during DENV infection.
Collapse
Affiliation(s)
- Lana Monteiro Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisa Beatriz Prestes
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle Premazzi Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, United States
| | | | - Yasmin Mucunã Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandro Silva da Costa
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Torres Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Barros Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Atalis A, Keenum MC, Pandey B, Beach A, Pradhan P, Vantucci C, Jain R, Hosten J, Smith C, Kramer L, Jimenez A, Ochoa MA, Frey D, Roy K. Nanoparticle-delivered TLR4 and RIG-I agonists enhance immune response to SARS-CoV-2 subunit vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.01.31.478507. [PMID: 35132413 PMCID: PMC8820660 DOI: 10.1101/2022.01.31.478507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Despite recent success in vaccinating populations against SARS-CoV-2, concerns about immunity duration, continued efficacy against emerging variants, protection from infection and transmission, and worldwide vaccine availability, remain. Although mRNA, pDNA, and viral-vector based vaccines are being administered, no protein subunit-based SARS-CoV-2 vaccine is approved. Molecular adjuvants targeting pathogen-recognition receptors (PRRs) on antigen-presenting cells (APCs) could improve and broaden the efficacy and durability of vaccine responses. Native SARS-CoV-2 infection stimulate various PRRs, including toll-like receptors (TLRs) and retinoic-acid-inducible gene I-like receptors (RIG-I). We hypothesized that targeting the same PRRs using adjuvants on nanoparticles along with a stabilized spike (S) protein antigen could provide broad and efficient immune responses. Formulations targeting TLR4 (MPLA), TLR7/8 (R848), TLR9 (CpG), and RIG-I (PUUC) delivered on degradable polymer-nanoparticles (NPs) were combined with the S1 subunit of S protein and assessed in vitro with isogeneic mixed lymphocyte reactions (iso-MLRs). For in vivo studies, the adjuvanted nanoparticles were combined with stabilized S protein and assessed using intranasal and intramuscular prime-boost vaccination models in mice. Combination NP-adjuvants targeting both TLR and RIG-I (MPLA+PUUC, CpG+PUUC, or R848+PUUC) differentially increased proinflammatory cytokine secretion (IL-1β, IL-12p70, IL-27, IFN-β) by APCs cultured in vitro, and induced differential T cell proliferation. When delivered intranasally, MPLA+PUUC NPs enhanced local CD4+CD44+ activated memory T cell responses while MPLA NPs increased anti-S-protein-specific IgG and IgA in the lung. Following intramuscular delivery, PUUC-carrying NPs induced strong humoral immune responses, characterized by increases in anti-S-protein IgG and neutralizing antibody titers and germinal center B cell populations (GL7+ and BCL6+ B cells). MPLA+PUUC NPs further boosted S-protein-neutralizing antibody titers and T follicular helper cell populations in draining lymph nodes. These results suggest that SARS-CoV-2-mimicking adjuvants and subunit vaccines could lead to robust and unique route-specific adaptive immune responses and may provide additional tools against the pandemic.
Collapse
|
13
|
Meuren LM, Coelho SVA, de Arruda LB. Evaluation of DENV-Induced Endothelial Cell Permeability by Measurements of Transendothelial Electrical Resistance (TEER) and Extravasation of Proteins and Virus. Methods Mol Biol 2022; 2409:207-222. [PMID: 34709644 DOI: 10.1007/978-1-0716-1879-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
This chapter will discuss reliable and relatively easy and fast strategies to evaluate the integrity of endothelial cell monolayers when infected by dengue virus (DENV). Human brain microvascular endothelial cells (HBMEC) were exploited here as general model of vessel wall core, but it may also be used as an in vitro simplified model of blood brain barrier (BBB). The integrity of endothelial cells monolayer can be inferred using a transwell culture system by: (1) measuring transendothelial electrical resistance (TEER) using a Voltohmmeter; (2) analyzing the monolayer permeability to fluorescent-conjugated proteins and fluorimetric assay; (3) investigating virus extravasation by quantitative RT-PCR and plaque conventional assay. The rational to use those strategies is that vascular alterations are often observed during dengue infection, being associated to disease severity. The vasculature core consists of a barrier of endothelial cells, which are tightly adhered by the expression of adhesion molecules and tight junctions. This structure must be preserved in order to control the flux of cells and metabolites from the circulation to the tissues and to maintain vascular homeostasis. Therefore, experimental assays that allow evaluation of endothelial integrity can be useful platforms to further understand disease pathogenesis and screen pharmaceutical interventions to control vascular disturbance.
Collapse
Affiliation(s)
- Lana Monteiro Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Sharton Vinicius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
14
|
Nanaware N, Banerjee A, Mullick Bagchi S, Bagchi P, Mukherjee A. Dengue Virus Infection: A Tale of Viral Exploitations and Host Responses. Viruses 2021; 13:v13101967. [PMID: 34696397 PMCID: PMC8541669 DOI: 10.3390/v13101967] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Dengue is a mosquito-borne viral disease (arboviral) caused by the Dengue virus. It is one of the prominent public health problems in tropical and subtropical regions with no effective vaccines. Every year around 400 million people get infected by the Dengue virus, with a mortality rate of about 20% among the patients with severe dengue. The Dengue virus belongs to the Flaviviridae family, and it is an enveloped virus with positive-sense single-stranded RNA as the genetic material. Studies of the infection cycle of this virus revealed potential host targets important for the virus replication cycle. Here in this review article, we will be discussing different stages of the Dengue virus infection cycle inside mammalian host cells and how host proteins are exploited by the virus in the course of infection as well as how the host counteracts the virus by eliciting different antiviral responses.
Collapse
Affiliation(s)
- Nikita Nanaware
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (N.N.); (A.B.)
| | - Anwesha Banerjee
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (N.N.); (A.B.)
| | | | - Parikshit Bagchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Correspondence: or (P.B.); or (A.M.)
| | - Anupam Mukherjee
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (N.N.); (A.B.)
- Correspondence: or (P.B.); or (A.M.)
| |
Collapse
|
15
|
Bakoa F, Préhaud C, Beauclair G, Chazal M, Mantel N, Lafon M, Jouvenet N. Genomic diversity contributes to the neuroinvasiveness of the Yellow fever French neurotropic vaccine. NPJ Vaccines 2021; 6:64. [PMID: 33903598 PMCID: PMC8076279 DOI: 10.1038/s41541-021-00318-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/15/2021] [Indexed: 02/02/2023] Open
Abstract
Mass vaccination with the live attenuated vaccine YF-17D is the current way to prevent infection with Yellow fever virus (YFV). However, 0.000012-0.00002% of vaccinated patients develop post-vaccination neurological syndrome (YEL-AND). Understanding the factors responsible for neuroinvasion, neurotropism, and neurovirulence of the vaccine is critical for improving its biosafety. The YF-FNV vaccine strain, known to be associated with a higher frequency of YEL-AND (0.3-0.4%) than YF-17D, is an excellent model to study vaccine neuroinvasiveness. We determined that neuroinvasiveness of YF-FNV occured both via infection and passage through human brain endothelial cells. Plaque purification and next generation sequencing (NGS) identified several neuroinvasive variants. Their neuroinvasiveness was not higher than that of YF-FNV. However, rebuilding the YF-FNV population diversity from a set of isolated YF-FNV-N variants restored the original neuroinvasive phenotype of YF-FNV. Therefore, we conclude that viral population diversity is a critical factor for YFV vaccine neuroinvasiveness.
Collapse
Affiliation(s)
- Florian Bakoa
- Unité de Neuroimmunologie Virale, Institut Pasteur, Paris, France
- Research and External Innovation Department, Sanofi Pasteur, Marcy L'Etoile, France
- Sorbonne Université, Collège doctoral, Paris, France
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France
| | | | - Guillaume Beauclair
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France
- Institut de Biologie Intégrative de la Cellule, UMR9198, Équipe Autophagie et Immunité Antivirale, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Maxime Chazal
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France
| | - Nathalie Mantel
- Research and External Innovation Department, Sanofi Pasteur, Marcy L'Etoile, France
| | - Monique Lafon
- Unité de Neuroimmunologie Virale, Institut Pasteur, Paris, France.
| | - Nolwenn Jouvenet
- Unité de Signalisation Antivirale, CNRS UMR 3569, Institut Pasteur, Paris, France.
| |
Collapse
|
16
|
Tembusu Virus entering the central nervous system caused nonsuppurative encephalitis without disrupting the blood-brain barrier. J Virol 2021; 95:JVI.02191-20. [PMID: 33472933 PMCID: PMC8092698 DOI: 10.1128/jvi.02191-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tembusu Virus (TMUV) is an emerging and re-emerging zoonotic pathogen that adversely affects poultry industry in recent years. TMUV disease is characterized by nonsuppurative encephalitis in ducklings. The duckling infection model was established to study the mechanism of TMUV crossing the blood-brain barrier (BBB) into the central nervous system (CNS). Here, we showed that no obvious clinical symptoms and enhancement of BBB permeability occurred at the early stage of infection (3∼5 dpi). While simultaneously virus particles were observed by transmission electron microscopy in the brain, inducing the accumulation of inflammatory cytokines. Neurological symptoms and disruption of BBB appeared at the intermediate stage of infection (7∼9 dpi). It was confirmed that TMUV could survive and propagate in brain microvascular endothelial cells (BMECs), but did not affect the permeability of BBB in vivo and in vitro at an early date. In conclusion, TMUV enters the CNS then causes encephalitis, and finally destruct the BBB, which may be due to the direct effect of TMUV on BMECs and the subsequent response of "inflammatory storm".IMPORTANCE The TMUV disease has caused huge losses to the poultry industry in Asia, which is potentially harmful to public health. Neurological symptoms and their sequelae are the main characters of this disease. However, the mechanism of how this virus enters the brain and causes encephalitis is unclear. In this study, we confirmed that the virus entered the CNS and then massively destroyed BBB and the BBB damage was closely associated with the subsequent outbreak of inflammation. TMUV may enter the CNS through the transcellular and "Trojan horse" pathways. These findings can fill the knowledge gap in the pathogenesis of TMUV-infected poultry and be benefit for the treatment of TMUV disease. What's more, TMUV is a representative to study the infection of avian flavivirus. Therefore, our studies have significances both for understanding of the full scope of mechanisms of TMUV and other flavivirus infection, and conceivably, for therapeutics.
Collapse
|
17
|
Coelho SVA, Rust NM, Vellasco L, Papa MP, Pereira ASG, da Silva Palazzo MF, Juliano MA, Costa SM, Alves AMB, Cordeiro MT, Marques ETA, Scharfstein J, de Arruda LB. Contact System Activation in Plasma from Dengue Patients Might Harness Endothelial Virus Replication through the Signaling of Bradykinin Receptors. Pharmaceuticals (Basel) 2021; 14:ph14010056. [PMID: 33445640 PMCID: PMC7827195 DOI: 10.3390/ph14010056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Since exacerbated inflammation and microvascular leakage are hallmarks of dengue virus (DENV) infection, here we interrogated whether systemic activation of the contact/kallikrein-kinin system (KKS) might hamper endothelial function. In vitro assays showed that dextran sulfate, a potent contact activator, failed to generate appreciable levels of activated plasma kallikrein (PKa) in the large majority of samples from a dengue cohort (n = 70), irrespective of severity of clinical symptoms. Impaired formation of PKa in dengue-plasmas correlated with the presence of cleaved Factor XII and high molecular weight kininogen (HK), suggesting that the prothrombogenic contact system is frequently triggered during the course of infection. Using two pathogenic arboviruses, DENV or Zika virus (ZIKV), we then asked whether exogenous BK could influence the outcome of infection of human brain microvascular endothelial cells (HBMECs). Unlike the unresponsive phenotype of Zika-infected HBMECs, we found that BK, acting via B2R, vigorously stimulated DENV-2 replication by reverting nitric oxide-driven apoptosis of endothelial cells. Using the mouse model of cerebral dengue infection, we next demonstrated that B2R targeting by icatibant decreased viral load in brain tissues. In summary, our study suggests that contact/KKS activation followed by BK-induced enhancement of DENV replication in the endothelium may underlie microvascular pathology in dengue.
Collapse
Affiliation(s)
- Sharton V. A. Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Naiara M. Rust
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Lucas Vellasco
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Michelle P. Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Aline S. G. Pereira
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Matheus Ferreira da Silva Palazzo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Maria Aparecida Juliano
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
| | - Simone M. Costa
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (S.M.C.); (A.M.B.A.)
| | - Ada M. B. Alves
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (S.M.C.); (A.M.B.A.)
| | - Marli T. Cordeiro
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Recife 50740-465, Brazil; (M.T.C.); (E.T.A.M.)
| | - Ernesto T. A. Marques
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Recife 50740-465, Brazil; (M.T.C.); (E.T.A.M.)
- Department of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Júlio Scharfstein
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
- Correspondence: (J.S.); (L.B.d.A.)
| | - Luciana B. de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
- Correspondence: (J.S.); (L.B.d.A.)
| |
Collapse
|
18
|
Xu X, Miao J, Shao Q, Gao Y, Hong L. Apigenin suppresses influenza A virus-induced RIG-I activation and viral replication. J Med Virol 2020; 92:3057-3066. [PMID: 32776519 DOI: 10.1002/jmv.26403] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/27/2020] [Indexed: 02/04/2023]
Abstract
Apigenin is a flavonoid of low toxicity and multiple beneficial bioactivities, including the properties of antitumor, antioxidant, anti-inflammatory, and antiviral activities. However, the effects of Apigenin on influenza virus infection remain poorly understood. Thus, the aim of this study is to investigate the effect of Apigenin on influenza A virus (IAV)-induced inflammation and viral replication. This study demonstrated that Apigenin treatment significantly suppressed IAV-induced upregulation of retinoic acid-inducible gene-I (RIG-I) expression, as well as the production of proinflammatory cytokines and interferons (IFN-β and IFN-λ1). Meanwhile, Apigenin also protected cells from IAV-induced cell death. In addition, Apigenin specifically inhibited the activation of RIG-I signaling via promoting the ubiquitin-mediated degradation of RIG-I, which may cause by the disrupting its interaction with heat shock protein 90α. Interestingly, instead of enhancing viral replication due to the inhibitory effects of Apigenin on the activation of RIG-I and expression of IFNs, Apigenin inhibited IAV replication in vitro. Further study demonstrated that Apigenin inhibited the influenza viral neuraminidase (NA) activity. Thus, Apigenin may serve as a promising supplementary approach for treatment of influenza because it protected cells from IAV-induced cell death and inhibited viral NA activity to suppress viral replication.
Collapse
Affiliation(s)
- Xiaohong Xu
- Department of Pediatrics, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jing Miao
- Department of Pediatrics, Binzhou People's Hospital, Binzhou, China
| | - Qingliang Shao
- Department of Pediatrics, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yue Gao
- Department of Pediatrics, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Luojia Hong
- Department of Hematology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Begum F, Das S, Mukherjee D, Mal S, Ray U. Insight into the Tropism of Dengue Virus in Humans. Viruses 2019; 11:v11121136. [PMID: 31835302 PMCID: PMC6950149 DOI: 10.3390/v11121136] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
In tropical and subtropical zones, arboviruses are among the major threats to human life, affecting a large number of populations with serious diseases. Worldwide, over three hundred million people are infected with dengue virus (DENV) every year as per the World Health Organization (WHO). DENV-mediated disease severity ranges from a mild fever to hemorrhagic fever and shock syndrome. Patients suffering from severe infection might experience multi-organ failure, cardiomyopathy and even encephalopathy, further complicating the disease pathogenesis. In life-threatening cases, DENV has been reported to affect almost all organs of the human body. In this review, we discuss the organ tropism of DENV in humans in depth as detected in various autopsy studies. Keeping in mind the fact that there is currently no DENV-specific antiviral, it is of utmost importance to achieve a vivid picture of the susceptible cells in humans which might help in designing antivirals against DENV, especially targeting those tissues in which infection might lead to life-threatening conditions.
Collapse
Affiliation(s)
- Feroza Begum
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sandeepan Das
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debica Mukherjee
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sweety Mal
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
| | - Upasana Ray
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata700032, India; (F.B.); (S.D.); (D.M.); (S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Correspondence: ; Tel.: +91-978-187-8333
| |
Collapse
|
20
|
The Interplay between Dengue Virus and the Human Innate Immune System: A Game of Hide and Seek. Vaccines (Basel) 2019; 7:vaccines7040145. [PMID: 31658677 PMCID: PMC6963221 DOI: 10.3390/vaccines7040145] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
With 40% of the world population at risk, infections with dengue virus (DENV) constitute a serious threat to public health. While there is no antiviral therapy available against this potentially lethal disease, the efficacy of the only approved vaccine is not optimal and its safety has been recently questioned. In order to develop better vaccines based on attenuated and/or chimeric viruses, one must consider how the human immune system is engaged during DENV infection. The activation of the innate immunity through the detection of viruses by cellular sensors is the first line of defence against those pathogens. This triggers a cascade of events which establishes an antiviral state at the cell level and leads to a global immunological response. However, DENV has evolved to interfere with the innate immune signalling at multiple levels, hence dampening antiviral responses and favouring viral replication and dissemination. This review elaborates on the interplay between DENV and the innate immune system. A special focus is given on the viral countermeasure mechanisms reported over the last decade which should be taken into consideration during vaccine development.
Collapse
|
21
|
Zhou B, Liang X, Feng Q, Li J, Pan X, Xie P, Jiang Z, Yang Z. Ergosterol peroxide suppresses influenza A virus-induced pro-inflammatory response and apoptosis by blocking RIG-I signaling. Eur J Pharmacol 2019; 860:172543. [PMID: 31323223 DOI: 10.1016/j.ejphar.2019.172543] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/06/2019] [Accepted: 07/15/2019] [Indexed: 12/25/2022]
Abstract
Ergosterol peroxide has been shown to exhibit anti-tumor, antioxidant and anti-bacterial properties. However, the effects of ergosterol peroxide isolated from the herbal Baphicacanthus cusia root on influenza virus infection remain poorly understood. In the present study, ergosterol peroxide (compound 22) was obtained from the B. cusia root and subjected to investigation regarding its immunoregulatory effect on influenza A virus (IAV)-induced inflammation in A549 human alveolar epithelial cells. The structure of compound 22 isolated from B. cusia root. was elucidated by NMR analyses. Structure determination showed that the chemical structure of compound 22 closely resembles that of ergosterol peroxide. We observed that ergosterol peroxide treatment significantly suppressed IAV-induced upregulation of RIG-I expression. Additionally, ergosterol peroxide inhibited the activation of RIG-I downstream signaling pathways, including p38 MAP kinase and NF-κB, which ultimately resulted in the reduced production of an array of pro-inflammatory mediators and interferons (IFN-β and IFN-λ1). Interestingly, inhibitory effects of ergosterol peroxide on the expression of IFNs did not affect the expression of antiviral effectors or enhance viral replication. On the other hand, ergosterol peroxide effectively abolished the amplified production of pro-inflammatory mediators in cells pretreated with IFN-β (500 ng/ml) prior to IAV infection. Moreover, Annexin V and Hoechst 33258 staining revealed that increased apoptosis of IAV-infected cells was reversed by the presence of ergosterol peroxide. Our findings suggest that ergosterol peroxide from the B. cusia root suppressed IAV-associated inflammation and apoptosis via blocking RIG-I signaling, which may serve as a supplementary approach to the treatment of influenza.
Collapse
Affiliation(s)
- Beixian Zhou
- Department of Pharmacy, The People's Hospital of Gaozhou, Gaozhou, Guangdong, 525200, China
| | - Xiaoli Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qitong Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China
| | - Jing Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xiping Pan
- Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Peifang Xie
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Zhihong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China.
| |
Collapse
|
22
|
Rath CT, Schnellrath LC, Damaso CR, de Arruda LB, Vasconcelos PFDC, Gomes C, Laurenti MD, Calegari Silva TC, Vivarini ÁDC, Fasel N, Pereira RMS, Lopes UG. Amazonian Phlebovirus (Bunyaviridae) potentiates the infection of Leishmania (Leishmania) amazonensis: Role of the PKR/IFN1/IL-10 axis. PLoS Negl Trop Dis 2019; 13:e0007500. [PMID: 31216268 PMCID: PMC6602282 DOI: 10.1371/journal.pntd.0007500] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 07/01/2019] [Accepted: 05/30/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Leishmania parasites are transmitted to vertebrate hosts by phlebotomine sandflies and, in humans, may cause tegumentary or visceral leishmaniasis. The role of PKR (dsRNA activated kinase) and Toll-like receptor 3 (TLR3) activation in the control of Leishmania infection highlights the importance of the engagement of RNA sensors, which are usually involved in the antiviral cell response, in the fate of parasitism by Leishmania. We tested the hypothesis that Phlebovirus, a subgroup of the Bunyaviridae, transmitted by sandflies, would interfere with Leishmania infection. METHODOLOGY/PRINCIPAL FINDINGS We tested two Phlebovirus isolates, Icoaraci and Pacui, from the rodents Nectomys sp. and Oryzomys sp., respectively, both natural sylvatic reservoir of Leishmania (Leishmania) amazonensis from the Amazon region. Phlebovirus coinfection with L. (L.) amazonensis in murine macrophages led to increased intracellular growth of L. (L.) amazonensis. Further studies with Icoaraci coinfection revealed the requirement of the PKR/IFN1 axis on the exacerbation of the parasite infection. L. (L.) amazonensis and Phlebovirus coinfection potentiated PKR activation and synergistically induced the expression of IFNβ and IL-10. Importantly, in vivo coinfection of C57BL/6 mice corroborated the in vitro data. The exacerbation effect of RNA virus on parasite infection may be specific because coinfection with dengue virus (DENV2) exerted the opposite effect on parasite load. CONCLUSIONS Altogether, our data suggest that coinfections with specific RNA viruses shared by vectors or reservoirs of Leishmania may enhance and sustain the activation of host cellular RNA sensors, resulting in aggravation of the parasite infection. The present work highlights new perspectives for the investigation of antiviral pathways as important modulators of protozoan infections.
Collapse
Affiliation(s)
- Carolina Torturella Rath
- Laboratory of Molecular Parasitology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laila Castro Schnellrath
- Laboratory of Molecular Biology of Virus, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarissa R. Damaso
- Laboratory of Molecular Biology of Virus, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Barros de Arruda
- Laboratório de Genética e Imunologia das Infecções Virais, Departamento de Virologia, Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Claudia Gomes
- Department of Pathology, Medical School, University of São Paulo, Brazil
| | | | - Teresa Cristina Calegari Silva
- Laboratory of Molecular Parasitology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Áislan de Carvalho Vivarini
- Laboratory of Molecular Parasitology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Switzerland
| | - Renata Meirelles Santos Pereira
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (RMSP); (UGL)
| | - Ulisses Gazos Lopes
- Laboratory of Molecular Parasitology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (RMSP); (UGL)
| |
Collapse
|
23
|
Klein RS, Garber C, Funk KE, Salimi H, Soung A, Kanmogne M, Manivasagam S, Agner S, Cain M. Neuroinflammation During RNA Viral Infections. Annu Rev Immunol 2019; 37:73-95. [PMID: 31026414 PMCID: PMC6731125 DOI: 10.1146/annurev-immunol-042718-041417] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neurotropic RNA viruses continue to emerge and are increasingly linked to diseases of the central nervous system (CNS) despite viral clearance. Indeed, the overall mortality of viral encephalitis in immunocompetent individuals is low, suggesting efficient mechanisms of virologic control within the CNS. Both immune and neural cells participate in this process, which requires extensive innate immune signaling between resident and infiltrating cells, including microglia and monocytes, that regulate the effector functions of antiviral T and B cells as they gain access to CNS compartments. While these interactions promote viral clearance via mainly neuroprotective mechanisms, they may also promote neuropathology and, in some cases, induce persistent alterations in CNS physiology and function that manifest as neurologic and psychiatric diseases. This review discusses mechanisms of RNA virus clearance and neurotoxicity during viral encephalitis with a focus on the cytokines essential for immune and neural cell inflammatory responses and interactions. Understanding neuroimmune communications in the setting of viral infections is essential for the development of treatments that augment neuroprotective processes while limiting ongoing immunopathological processes that cause ongoing CNS disease.
Collapse
Affiliation(s)
- Robyn S Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Charise Garber
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Kristen E Funk
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Hamid Salimi
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Allison Soung
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Marlene Kanmogne
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Sindhu Manivasagam
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Shannon Agner
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Matthew Cain
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| |
Collapse
|
24
|
Mustafá YM, Meuren LM, Coelho SVA, de Arruda LB. Pathways Exploited by Flaviviruses to Counteract the Blood-Brain Barrier and Invade the Central Nervous System. Front Microbiol 2019; 10:525. [PMID: 30984122 PMCID: PMC6447710 DOI: 10.3389/fmicb.2019.00525] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
Human infection by different flaviviruses may cause severe neurologic syndromes, through pathogenic mechanisms that are still largely unknown. Japanese encephalitis virus (JEV), West Nile virus (WNV), Zika virus (ZIKV), yellow fever virus (YFV), dengue virus (DENV), and tick-borne encephalitis virus (TBEV) are believed to reach the central nervous system by a hematogenous route, upon crossing the blood-brain barrier. Although the disruption of BBB during flavivirus infection has been largely evidenced in experimental models, the relevance of BBB breakdown for virus entering the brain was not completely elucidated. In vitro models of BBB had demonstrated that these viruses replicated in brain microvascular endothelial cells (BMECs), which induced downregulation of tight junction proteins and increased the permeability of the barrier. Other reports demonstrated that infection of BMECs allowed the basolateral release of infectious particles, without a remarkable cytopathic effect, what might be sufficient for virus invasion. Virus replication and activation of other cells associated to the BBB, mostly astrocytes and microglia, were also reported to affect the endothelial barrier permeability. This event might occur simultaneously or after BMECs infection, being a secondary effect leading to BBB disruption. Importantly, activation of BMECs, astrocytes, and microglia by flaviviruses was associated to the expression and secretion of inflammatory mediators, which are believed to recruit leukocytes to the CNS. The leukocyte infiltrate could further mediate viral invasion through a Trojan horse mechanism and might contribute to BBB breakdown and to neurological alterations. This review discussed the previous studies regarding in vitro and in vivo models of JEV, WNV, ZIKV, YFV, DENV, and TBEV infection and addressed the pathways for BBB overcome and invasion of the CNS described for each virus infection, aiming to increment the knowledge and stimulate further discussion about the role of BBB in the neuropathogenesis of flavivirus infection.
Collapse
Affiliation(s)
- Yasmin Mucunã Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lana Monteiro Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Sharton Vinícius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Pais TF, Penha-Gonçalves C. Brain Endothelium: The "Innate Immunity Response Hypothesis" in Cerebral Malaria Pathogenesis. Front Immunol 2019; 9:3100. [PMID: 30761156 PMCID: PMC6361776 DOI: 10.3389/fimmu.2018.03100] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/14/2018] [Indexed: 01/04/2023] Open
Abstract
Cerebral malaria (CM) is a life-threatening neurological syndrome caused by Plasmodium falciparum infection afflicting mainly children in Africa. Current pathogenesis models implicate parasite and host-derived factors in impairing brain vascular endothelium (BVE) integrity. Sequestration of Plasmodium-infected red blood cells (iRBCs) in brain microvessels is a hallmark of CM pathology. However, the precise mechanisms driving loss of blood-brain barrier (BBB) function with consequent brain injury are still unsettled and it is plausible that distinct pathophysiology trajectories are involved. Studies in humans and in the mouse model of CM indicate that inflammatory reactions intertwined with microcirculatory and coagulation disturbances induce alterations in vascular permeability and impair BBB integrity. Yet, the role of BVE as initiator of immune responses against parasite molecules and iRBCs is largely unexplored. Brain endothelial cells express pattern recognition receptors (PRR) and are privileged sensors of blood-borne infections. Here, we focus on the hypothesis that innate responses initiated by BVE and subsequent interactions with immune cells are critical to trigger local effector immune functions and induce BBB damage. Uncovering mechanisms of BVE involvement in sensing Plasmodium infection, recruiting of immune cells and directing immune effector functions could reveal pharmacological targets to promote BBB protection with potential applications in CM clinical management.
Collapse
|
26
|
Li G, Tian Y, Chen L, Shen J, Tao Z, Zeng T, Xu J, Lu L. Cloning, expression, and bioinformatics analysis of a putative pigeon retinoid acid-inducible gene-I. CANADIAN JOURNAL OF ANIMAL SCIENCE 2018. [DOI: 10.1139/cjas-2017-0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Retinoid acid-inducible gene-I (RIG-I) is a major cytoplasmic RNA sensor, playing an essential role in detecting viral RNA and triggering antiviral innate immune responses. The objective of the present study was to characterize the structure and expression of the RIG-I gene in pigeons. The pigeon RIG-I (piRIG-I) was cloned from splenic lymphocytes of pigeons using reverse transcription polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends. The cDNA of piRIG-I contains a 147 bp 5′-untranslated regions (UTRs), a 2787 bp open reading frame, and 2962 bp 3′-UTRs. Based on this sequence, the encoded piRIG-I protein is predicted to consist of 928 amino acids, and it has conserved domains typical of RIG-I-like receptors (RLRs) including two tandem arranged N-terminal caspase recruitment domains, a domain with the signature of DExD/H box helicase (helicase domain), and a C-terminal repression domain similar to finch RIG-I, duck RIG-I, goose RIG-I, human RIG-I, and mouse RIG-I. The piRIG-I shows 82.1%, 78.6%, and 78.2% amino acid sequence identity with previously described finch RIG-I, duck RIG-I, and goose RIG-I, respectively, and 49.7%–53.8% sequence identity with mammalian homologs. Quantitative RT-PCR (qRT-PCR) analysis indicated that the piRIG-I mRNA is scarcely detected in healthy tissues, and it is strongly expressed in the thymus gland, kidney, spleen, and bursa of Fabricius. These findings lay the foundation for further research on the function and mechanism of avian RIG-I in innate immune response related to vaccinations and infectious diseases in the pigeon.
Collapse
Affiliation(s)
- Guoqin Li
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| | - Yong Tian
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| | - Li Chen
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| | - Junda Shen
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| | - Zhengrong Tao
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| | - Tao Zeng
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| | - Jian Xu
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| | - Lizhi Lu
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
- Institute of Animal Science and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, 198 Shiqiao Road, Hangzhou 310021, People’s Republic of China
| |
Collapse
|
27
|
Konradt C, Hunter CA. Pathogen interactions with endothelial cells and the induction of innate and adaptive immunity. Eur J Immunol 2018; 48:1607-1620. [PMID: 30160302 DOI: 10.1002/eji.201646789] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 07/24/2018] [Accepted: 08/23/2018] [Indexed: 12/28/2022]
Abstract
There are over 10 trillion endothelial cells (EC) that line the vasculature of the human body. These cells not only have specialized functions in the maintenance of homeostasis within the circulation and various tissues but they also have a major role in immune function. EC also represent an important replicative niche for a subset of viral, bacterial, and parasitic organisms that are present in the blood or lymph; however, there are major gaps in our knowledge regarding how pathogens interact with EC and how this influences disease outcome. In this article, we review the literature on EC-pathogen interactions and their role in innate and adaptive mechanisms of resistance to infection and highlight opportunities to address prominent knowledge gaps.
Collapse
Affiliation(s)
- Christoph Konradt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
28
|
Chazal M, Beauclair G, Gracias S, Najburg V, Simon-Lorière E, Tangy F, Komarova AV, Jouvenet N. RIG-I Recognizes the 5' Region of Dengue and Zika Virus Genomes. Cell Rep 2018; 24:320-328. [PMID: 29996094 DOI: 10.1016/j.celrep.2018.06.047] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/21/2018] [Accepted: 06/08/2018] [Indexed: 12/24/2022] Open
Abstract
The flavivirus genus comprises major human pathogens, such as Dengue (DENV) and Zika (ZIKV) viruses. RIG-I and MDA5 are key cytoplasmic pathogen recognition receptors that are implicated in detecting viral RNAs. Here, we show that RNAs that co-purified with RIG-I during DENV infection are immuno-stimulatory, whereas RNAs bound to MDA5 are not. An affinity purification method combined with next-generation sequencing (NGS) revealed that the 5' region of the DENV genome is recognized by RIG-I. No DENV RNA was bound to MDA5. In vitro production of fragments of the DENV genome confirmed the NGS data and revealed that the 5' end of the genome, when bearing 5'-triphosphates, is the RIG-I ligand. The 5' region of the ZIKV genome is also a RIG-I agonist. We propose that RIG-I binds to the highly structured and conserved 5' region of flavivirus nascent transcripts before capping and that this mechanism leads to interferon secretion by infected cells.
Collapse
Affiliation(s)
- Maxime Chazal
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France; Unité de Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris 75015, France; CNRS UMR2000 Génomique Évolutive, Modélisation et Santé, Institut Pasteur, Paris 75015, France
| | - Guillaume Beauclair
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Ségolène Gracias
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Valérie Najburg
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Etienne Simon-Lorière
- Unité de Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris 75015, France; CNRS UMR2000 Génomique Évolutive, Modélisation et Santé, Institut Pasteur, Paris 75015, France
| | - Frédéric Tangy
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Anastassia V Komarova
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France.
| | - Nolwenn Jouvenet
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France.
| |
Collapse
|
29
|
Flaviviral RNA Structures and Their Role in Replication and Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1062:45-62. [PMID: 29845524 DOI: 10.1007/978-981-10-8727-1_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
More than simple vectors of genetic information, flaviviral RNAs have emerged as critical regulators of the virus life cycle. Viral RNAs regulate interactions with viral and cellular proteins in both, mosquito and mammalian hosts to ultimately influence processes as diverse as RNA replication, translation, packaging or pathogenicity. In this chapter, we will review the current knowledge of the role of sequence and structures in the flaviviral RNA in viral propagation and interaction with the host cell. We will also cover the increasing body of evidence linking viral non-coding RNAs with pathogenicity, host immunity and epidemic potential.
Collapse
|
30
|
Boff D, Fagundes CT, Russo RC, Amaral FA. Innate Immunity and Inflammation: The Molecular Mechanisms Governing the Cross-Talk Between Innate Immune and Endothelial Cells. IMMUNOPHARMACOLOGY AND INFLAMMATION 2018:33-56. [DOI: 10.1007/978-3-319-77658-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Papa MP, Meuren LM, Coelho SVA, Lucas CGDO, Mustafá YM, Lemos Matassoli F, Silveira PP, Frost PS, Pezzuto P, Ribeiro MR, Tanuri A, Nogueira ML, Campanati L, Bozza MT, Paula Neto HA, Pimentel-Coelho PM, Figueiredo CP, de Aguiar RS, de Arruda LB. Zika Virus Infects, Activates, and Crosses Brain Microvascular Endothelial Cells, without Barrier Disruption. Front Microbiol 2017; 8:2557. [PMID: 29312238 PMCID: PMC5743735 DOI: 10.3389/fmicb.2017.02557] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 12/08/2017] [Indexed: 12/19/2022] Open
Abstract
Zika virus (ZIKV) has been associated to central nervous system (CNS) harm, and virus was detected in the brain and cerebrospinal fluids of microcephaly and meningoencephalitis cases. However, the mechanism by which the virus reaches the CNS is unclear. Here, we addressed the effects of ZIKV replication in human brain microvascular endothelial cells (HBMECs), as an in vitro model of blood brain barrier (BBB), and evaluated virus extravasation and BBB integrity in an in vivo mouse experimental model. HBMECs were productively infected by African and Brazilian ZIKV strains (ZIKVMR766 and ZIKVPE243), which induce increased production of type I and type III IFN, inflammatory cytokines and chemokines. Infection with ZIKVMR766 promoted earlier cellular death, in comparison to ZIKVPE243, but infection with either strain did not result in enhanced endothelial permeability. Despite the maintenance of endothelial integrity, infectious virus particles crossed the monolayer by endocytosis/exocytosis-dependent replication pathway or by transcytosis. Remarkably, both viruses' strains infected IFNAR deficient mice, with high viral load being detected in the brains, without BBB disruption, which was only detected at later time points after infection. These data suggest that ZIKV infects and activates endothelial cells, and might reach the CNS through basolateral release, transcytosis or transinfection processes. These findings further improve the current knowledge regarding ZIKV dissemination pathways.
Collapse
Affiliation(s)
- Michelle P. Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lana M. Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sharton V. A. Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina G. de Oliveira Lucas
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yasmin M. Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavio Lemos Matassoli
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola P. Silveira
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula S. Frost
- Núcleo de Neurociências da Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Pezzuto
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milene R. Ribeiro
- Laboratório de Pesquisas em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauricio L. Nogueira
- Laboratório de Pesquisas em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
| | - Loraine Campanati
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor A. Paula Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro M. Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia P. Figueiredo
- Núcleo de Neurociências da Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato S. de Aguiar
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana B. de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Interplay between dengue virus and Toll-like receptors, RIG-I/MDA5 and microRNAs: Implications for pathogenesis. Antiviral Res 2017; 147:47-57. [DOI: 10.1016/j.antiviral.2017.09.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 12/25/2022]
|
33
|
Li S, Nishikawa T, Kaneda Y. Inactivated Sendai virus particle upregulates cancer cell expression of intercellular adhesion molecule-1 and enhances natural killer cell sensitivity on cancer cells. Cancer Sci 2017; 108:2333-2341. [PMID: 28945328 PMCID: PMC5715349 DOI: 10.1111/cas.13408] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 09/17/2017] [Accepted: 09/21/2017] [Indexed: 12/31/2022] Open
Abstract
We have already reported that the inactivated Sendai virus (hemagglutinating virus of Japan; HVJ) envelope (HVJ‐E) has multiple anticancer effects, including induction of cancer‐selective cell death and activation of anticancer immunity. The HVJ‐E stimulates dendritic cells to produce cytokines and chemokines such as β‐interferon, interleukin‐6, chemokine (C‐C motif) ligand 5, and chemokine (C‐X‐C motif) ligand 10, which activate both CD8+ T cells and natural killer (NK) cells and recruit them to the tumor microenvironment. However, the effect of HVJ‐E on modulating the sensitivity of cancer cells to immune cell attack has yet to be investigated. In this study, we found that HVJ‐E induced the production of intercellular adhesion molecule‐1 (ICAM‐1, CD54), a ligand of lymphocyte function‐associated antigen 1, in several cancer cell lines through the activation of nuclear factor‐κB downstream of retinoic acid‐inducible gene I and the mitochondrial antiviral signaling pathway. The upregulation of ICAM‐1 on the surface of cancer cells increased the sensitivity of cancer cells to NK cells. Knocking out expression of ICAM‐1 in MDA‐MB‐231 cells using the CRISPR/Cas9 method significantly reduced the killing effect of NK cells on ICAM‐1‐depleted MDA‐MB‐231 cells. In addition, HVJ‐E suppressed tumor growth in MDA‐MB‐231 tumor‐bearing SCID mice, and the HVJ‐E antitumor effect was impaired when NK cells were depleted by treatment with the anti‐asialo GM1 antibody. Our findings suggest that HVJ‐E enhances NK cell sensitivity against cancer cells by increasing ICAM‐1 expression on the cancer cell surface.
Collapse
Affiliation(s)
- Simin Li
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomoyuki Nishikawa
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
34
|
Sprokholt JK, Kaptein TM, van Hamme JL, Overmars RJ, Gringhuis SI, Geijtenbeek TBH. RIG-I-like Receptor Triggering by Dengue Virus Drives Dendritic Cell Immune Activation and T H1 Differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:4764-4771. [PMID: 28507028 DOI: 10.4049/jimmunol.1602121] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/13/2017] [Indexed: 01/07/2023]
Abstract
Dengue virus (DENV) causes 400 million infections annually and is one of several viruses that can cause viral hemorrhagic fever, which is characterized by uncontrolled immune activation resulting in high fever and internal bleeding. Although the underlying mechanisms are unknown, massive cytokine secretion is thought to be involved. Dendritic cells (DCs) are the main target cells of DENV, and we investigated their role in DENV-induced cytokine production and adaptive immune responses. DENV infection induced DC maturation and secretion of IL-1β, IL-6, and TNF. Inhibition of DENV RNA replication abrogated these responses. Notably, silencing of RNA sensors RIG-I or MDA5 abrogated DC maturation, as well as cytokine responses by DENV-infected DCs. DC maturation was induced by type I IFN responses because inhibition of IFN-α/β receptor signaling abrogated DENV-induced DC maturation. Moreover, DENV infection of DCs resulted in CCL2, CCL3, and CCL4 expression, which was abrogated after RIG-I and MDA5 silencing. DCs play an essential role in TH cell differentiation, and we show that RIG-I and MDA5 triggering by DENV leads to TH1 polarization, which is characterized by high levels of IFN-γ. Notably, cytokines IL-6, TNF, and IFN-γ and chemokines CCL2, CCL3, and CCL4 have been associated with disease severity, endothelial dysfunction, and vasodilation. Therefore, we identified RIG-I and MDA5 as critical players in innate and adaptive immune responses against DENV, and targeting these receptors has the potential to decrease hemorrhagic fever in patients.
Collapse
Affiliation(s)
- Joris K Sprokholt
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; and
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - Tanja M Kaptein
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; and
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - John L van Hamme
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; and
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - Ronald J Overmars
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; and
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - Sonja I Gringhuis
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; and
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; and
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| |
Collapse
|
35
|
Non-Canonical Roles of Dengue Virus Non-Structural Proteins. Viruses 2017; 9:v9030042. [PMID: 28335410 PMCID: PMC5371797 DOI: 10.3390/v9030042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae family comprises a number of human pathogens, which, although sharing structural and functional features, cause diseases with very different outcomes. This can be explained by the plurality of functions exerted by the few proteins coded by viral genomes, with some of these functions shared among members of a same family, but others being unique for each virus species. These non-canonical functions probably have evolved independently and may serve as the base to the development of specific therapies for each of those diseases. Here it is discussed what is currently known about the non-canonical roles of dengue virus (DENV) non-structural proteins (NSPs), which may account for some of the effects specifically observed in DENV infection, but not in other members of the Flaviviridae family. This review explores how DENV NSPs contributes to the physiopathology of dengue, evasion from host immunity, metabolic changes, and redistribution of cellular components during infection.
Collapse
|
36
|
Sprokholt J, Helgers LC, Geijtenbeek TBH. Innate immune receptors drive dengue virus immune activation and disease. Future Virol 2017; 13:287-305. [PMID: 29937918 PMCID: PMC6004600 DOI: 10.2217/fvl-2017-0146] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/25/2018] [Indexed: 12/14/2022]
Abstract
Dengue is a worldwide disease with 400 million annual infections that can lead to septic shock and viral hemorrhagic fever with internal bleeding. These symptoms are the result of uncontrolled immune activation. Macrophages and dendritic cells are the main target of dengue virus (DENV) and the cellular source of cytokines associated with this immune activation. Macrophages and dendritic cells express several innate immune receptors that have been implicated in DENV immune activation, of which, CLEC5A, RIG-I and MDA5 are most important. Notably, activation of these receptors have profound effects on adaptive immune responses against DENV. This review will focus on how innate immune receptors drive DENV immune activation by inducing inflammatory cytokines and by activating adaptive immune responses.
Collapse
Affiliation(s)
- Joris Sprokholt
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, AMC, VUmc, Amsterdam, The Netherlands
| | - Leanne C Helgers
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, AMC, VUmc, Amsterdam, The Netherlands
| | - Teunis BH Geijtenbeek
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, AMC, VUmc, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Bustos-Arriaga J, Mita-Mendoza NK, Lopez-Gonzalez M, García-Cordero J, Juárez-Delgado FJ, Gromowski GD, Méndez-Cruz RA, Fairhurst RM, Whitehead SS, Cedillo-Barrón L. Soluble mediators produced by the crosstalk between microvascular endothelial cells and dengue-infected primary dermal fibroblasts inhibit dengue virus replication and increase leukocyte transmigration. Immunol Res 2016; 64:392-403. [PMID: 26130295 DOI: 10.1007/s12026-015-8675-8] [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: 11/26/2022]
Abstract
When dengue virus (DENV)-infected mosquitoes use their proboscis to probe into human skin during blood feeding, both saliva and virus are released. During this process, cells from the epidermis and dermis layers of the skin, along with small blood vessels, may get exposed to or infected with DENV. In these microenvironments of the skin, the presence of DENV initiates a complex interplay among the DENV-infected and non-infected neighboring cells at the initial bite site. Previous studies suggested that DENV-infected human dermal fibroblasts (HDFs) participate in the immune response against DENV by secreting soluble mediators of innate immunity. In the present study, we investigated whether DENV-infected HDFs activate human dermal microvascular endothelial cells (HDMECs) in co-cultures. Our results suggest that co-cultures of DENV-infected HDFs and HDMECs elicit soluble mediators that are sufficient to reduce viral replication, activate HDMECs, and induce leukocyte migration through HDMEC monolayers. These effects were partly dependent on HDF donor and DENV serotype, which may provide novel insights into the natural variation in host susceptibility to DENV disease.
Collapse
Affiliation(s)
- José Bustos-Arriaga
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neida K Mita-Mendoza
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Moises Lopez-Gonzalez
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico
| | - Julio García-Cordero
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico
| | | | - Gregory D Gromowski
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - René A Méndez-Cruz
- Laboratorio de Inmunología UMF, FES Iztacala, Universidad Autónoma de México, Mexico, Distrito Federal, Mexico
| | - Rick M Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leticia Cedillo-Barrón
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico.
| |
Collapse
|
38
|
Valadão ALC, Aguiar RS, de Arruda LB. Interplay between Inflammation and Cellular Stress Triggered by Flaviviridae Viruses. Front Microbiol 2016; 7:1233. [PMID: 27610098 PMCID: PMC4996823 DOI: 10.3389/fmicb.2016.01233] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae family comprises several human pathogens, including Dengue, Zika, Yellow Fever, West Nile, Japanese Encephalitis viruses, and Hepatitis C Virus. Those are enveloped, single-stranded positive sense RNA viruses, which replicate mostly in intracellular compartments associated to endoplasmic reticulum (ER) and Golgi complex. Virus replication results in abundant viral RNAs and proteins, which are recognized by cellular mechanisms evolved to prevent virus infection, resulting in inflammation and stress responses. Virus RNA molecules are sensed by Toll-like receptors (TLRs), RIG-I-like receptors (RIG-I and MDA5) and RNA-dependent protein kinases (PKR), inducing the production of inflammatory mediators and interferons. Simultaneously, the synthesis of virus RNA and proteins are distinguished in different compartments such as mitochondria, ER and cytoplasmic granules, triggering intracellular stress pathways, including oxidative stress, unfolded protein response pathway, and stress granules assembly. Here, we review the new findings that connect the inflammatory pathways to cellular stress sensors and the strategies of Flaviviridae members to counteract these cellular mechanisms and escape immune response.
Collapse
Affiliation(s)
- Ana L C Valadão
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Renato S Aguiar
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Luciana B de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
39
|
Schmid B, Rinas M, Ruggieri A, Acosta EG, Bartenschlager M, Reuter A, Fischl W, Harder N, Bergeest JP, Flossdorf M, Rohr K, Höfer T, Bartenschlager R. Live Cell Analysis and Mathematical Modeling Identify Determinants of Attenuation of Dengue Virus 2'-O-Methylation Mutant. PLoS Pathog 2015; 11:e1005345. [PMID: 26720415 PMCID: PMC4697809 DOI: 10.1371/journal.ppat.1005345] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 11/26/2015] [Indexed: 11/19/2022] Open
Abstract
Dengue virus (DENV) is the most common mosquito-transmitted virus infecting ~390 million people worldwide. In spite of this high medical relevance, neither a vaccine nor antiviral therapy is currently available. DENV elicits a strong interferon (IFN) response in infected cells, but at the same time actively counteracts IFN production and signaling. Although the kinetics of activation of this innate antiviral defense and the timing of viral counteraction critically determine the magnitude of infection and thus disease, quantitative and kinetic analyses are lacking and it remains poorly understood how DENV spreads in IFN-competent cell systems. To dissect the dynamics of replication versus antiviral defense at the single cell level, we generated a fully viable reporter DENV and host cells with authentic reporters for IFN-stimulated antiviral genes. We find that IFN controls DENV infection in a kinetically determined manner that at the single cell level is highly heterogeneous and stochastic. Even at high-dose, IFN does not fully protect all cells in the culture and, therefore, viral spread occurs even in the face of antiviral protection of naïve cells by IFN. By contrast, a vaccine candidate DENV mutant, which lacks 2’-O-methylation of viral RNA is profoundly attenuated in IFN-competent cells. Through mathematical modeling of time-resolved data and validation experiments we show that the primary determinant for attenuation is the accelerated kinetics of IFN production. This rapid induction triggered by mutant DENV precedes establishment of IFN-resistance in infected cells, thus causing a massive reduction of virus production rate. In contrast, accelerated protection of naïve cells by paracrine IFN action has negligible impact. In conclusion, these results show that attenuation of the 2’-O-methylation DENV mutant is primarily determined by kinetics of autocrine IFN action on infected cells. Dengue virus (DENV) infection is a global health problem for which no selective therapy or vaccine exists. The magnitude of infection critically depends on the induction kinetics of the interferon (IFN) response and the kinetics of viral countermeasures. Here we established a novel live cell imaging system to dissect the dynamics of this interplay. We find that IFN controls DENV infection in a kinetically determined manner. At the single cell level, the IFN response is highly heterogeneous and stochastic, likely accounting for viral spread in the presence of IFN. Mathematical modeling and validation experiments show that the kinetics of activation of the IFN response critically determines control of virus replication and spread. A vaccine candidate DENV mutant lacking 2’-O-methylation of viral RNA is profoundly attenuated in IFN-competent cells. This attenuation is primarily due to accelerated kinetics of IFN production acting on infected cells in an autocrine manner. In contrast, accelerated protection of naïve cells by paracrine IFN action has negligible impact. Thus, attenuation of the 2’-O-methylation DENV mutant is primarily determined by kinetics of autocrine IFN action on infected cells.
Collapse
Affiliation(s)
- Bianca Schmid
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Melanie Rinas
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
| | - Alessia Ruggieri
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Eliana Gisela Acosta
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Marie Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Antje Reuter
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Fischl
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Nathalie Harder
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
- Department of Bioinformatics and Functional Genomics, Biomedical Computer Vision Group, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Jan-Philip Bergeest
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
- Department of Bioinformatics and Functional Genomics, Biomedical Computer Vision Group, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Michael Flossdorf
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
| | - Karl Rohr
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
- Department of Bioinformatics and Functional Genomics, Biomedical Computer Vision Group, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
- * E-mail: (TH); (RB)
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
- * E-mail: (TH); (RB)
| |
Collapse
|
40
|
Dengue Virus-Induced Inflammation of the Endothelium and the Potential Roles of Sphingosine Kinase-1 and MicroRNAs. Mediators Inflamm 2015; 2015:509306. [PMID: 26609198 PMCID: PMC4644833 DOI: 10.1155/2015/509306] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/02/2015] [Accepted: 10/08/2015] [Indexed: 01/24/2023] Open
Abstract
One of the main pathogenic effects of severe dengue virus (DENV) infection is a vascular leak syndrome. There are no available antivirals or specific DENV treatments and without hospital support severe DENV infection can be life-threatening. The cause of the vascular leakage is permeability changes in the endothelial cells lining the vasculature that are brought about by elevated vasoactive cytokine and chemokines induced following DENV infection. The source of these altered cytokine and chemokines is traditionally believed to be from DENV-infected cells such as monocyte/macrophages and dendritic cells. Herein we discuss the evidence for the endothelium as an additional contributor to inflammatory and innate responses during DENV infection which may affect endothelial cell function, in particular the ability to maintain vascular integrity. Furthermore, we hypothesise roles for two factors, sphingosine kinase-1 and microRNAs (miRNAs), with a focus on several candidate miRNAs, which are known to control normal vascular function and inflammatory responses. Both of these factors may be potential therapeutic targets to regulate inflammation of the endothelium during DENV infection.
Collapse
|
41
|
Calvert JK, Helbig KJ, Dimasi D, Cockshell M, Beard MR, Pitson SM, Bonder CS, Carr JM. Dengue Virus Infection of Primary Endothelial Cells Induces Innate Immune Responses, Changes in Endothelial Cells Function and Is Restricted by Interferon-Stimulated Responses. J Interferon Cytokine Res 2015; 35:654-65. [PMID: 25902155 DOI: 10.1089/jir.2014.0195] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although endothelial cell (EC) infection is not widespread during dengue virus (DENV) infection in vivo, the endothelium is the site of the pathogenic effects seen in severe DENV disease. In this study, we investigated DENV infection of primary EC and defined factors that influence infection in this cell type. Consistent with in vivo findings where EC infection is infrequent, only 3%-15% of EC became productively DENV-2-infected in vitro. This low level infection could not be attributed to inhibition by heparin, EC donor variation, heterogeneity, or biological source. DENV-infection of EC was associated with induction of innate immune responses, including increased STAT1 protein, STAT1- phosphorylation, interferon (IFN)-β, OAS-1, IFIT-1/ISG56, and viperin mRNA. Antibody blocking of IFN-β inhibited the induction of OAS1, IFIT1/ISG56, and viperin while shRNA knockdown of viperin enhanced DENV-infection in EC. DENV-infection of EC resulted in increased activity of sphingosine kinase 1, a factor important in maintaining vascular integrity, and altered basal and stimulated changes in barrier integrity of DENV-infected EC monolayers. Thus, DENV productively infects only a small percentage of primary EC but this has a major influence on induction of IFN-β driven innate immune responses that can restrict infection while the EC themselves are functionally altered. These changes may have important consequences for the endothelium and are reflective of pathogenic changes associated with vascular leakage, as seen in DENV disease.
Collapse
Affiliation(s)
- Julie K Calvert
- 1 Microbiology and Infectious Diseases, School of Medicine, Flinders University , Adelaide, South Australia
| | - Karla J Helbig
- 2 School of Molecular and Biomedical Science, University of Adelaide , Adelaide, South Australia
| | - David Dimasi
- 3 Centre for Cancer Biology, University of South Australia and SA Pathology , Adelaide, South Australia
| | - Michaelia Cockshell
- 3 Centre for Cancer Biology, University of South Australia and SA Pathology , Adelaide, South Australia
| | - Michael R Beard
- 2 School of Molecular and Biomedical Science, University of Adelaide , Adelaide, South Australia.,3 Centre for Cancer Biology, University of South Australia and SA Pathology , Adelaide, South Australia
| | - Stuart M Pitson
- 3 Centre for Cancer Biology, University of South Australia and SA Pathology , Adelaide, South Australia
| | - Claudine S Bonder
- 3 Centre for Cancer Biology, University of South Australia and SA Pathology , Adelaide, South Australia
| | - Jillian M Carr
- 1 Microbiology and Infectious Diseases, School of Medicine, Flinders University , Adelaide, South Australia
| |
Collapse
|
42
|
Castillo Ramirez JA, Urcuqui-Inchima S. Dengue Virus Control of Type I IFN Responses: A History of Manipulation and Control. J Interferon Cytokine Res 2015; 35:421-30. [PMID: 25629430 DOI: 10.1089/jir.2014.0129] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The arthropod-borne diseases caused by dengue virus (DENV) are a major and emerging problem of public health worldwide. Infection with DENV causes a series of clinical manifestations ranging from mild flu syndrome to severe diseases that include hemorrhage and shock. It has been demonstrated that the innate immune response plays a key role in DENV pathogenesis. However, in recent years, it was shown that DENV evades the innate immune response by blocking type I interferon (IFN-I). It has been demonstrated that DENV can inhibit both the production and the signaling of IFN-I. The viral proteins, NS2A and NS3, inhibit IFN-I production by degrading cellular signaling molecules. In addition, the viral proteins, NS2A, NS4A, NS4B, and NS5, can inhibit IFN-I signaling by blocking the phosphorylation of the STAT1 and STAT2 molecules. Finally, NS5 mediates the degradation of STAT2 using the proteasome machinery. In this study, we briefly review the most recent insights regarding the IFN-I response to DENV infection and its implication for pathogenesis.
Collapse
Affiliation(s)
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA , Medellín, Colombia
| |
Collapse
|
43
|
Vervaeke P, Vermeire K, Liekens S. Endothelial dysfunction in dengue virus pathology. Rev Med Virol 2014; 25:50-67. [PMID: 25430853 DOI: 10.1002/rmv.1818] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/07/2014] [Accepted: 10/14/2014] [Indexed: 02/04/2023]
Abstract
Dengue virus (DENV) is a leading cause of illness and death, mainly in the (sub)tropics, where it causes dengue fever and/or the more serious diseases dengue hemorrhagic fever and dengue shock syndrome that are associated with changes in vascular permeability. Despite extensive research, the pathogenesis of DENV is still poorly understood and, although endothelial cells represent the primary fluid barrier of the blood vessels, the extent to which these cells contribute to DENV pathology is still under debate. The primary target cells for DENV are dendritic cells and monocytes/macrophages that release various chemokines and cytokines upon infection, which can activate the endothelium and are thought to play a major role in DENV-induced vascular permeability. However, recent studies indicate that DENV also replicates in endothelial cells and that DENV-infected endothelial cells may directly contribute to viremia, immune activation, vascular permeability and immune targeting of the endothelium. Also, the viral non-structural protein-1 and antibodies directed against this secreted protein have been reported to be involved in endothelial cell dysfunction. This review provides an extensive overview of the effects of DENV infection on endothelial cell physiology and barrier function.
Collapse
Affiliation(s)
- Peter Vervaeke
- KU Leuven, Rega Institute for Medical Research, Leuven, Belgium
| | | | | |
Collapse
|
44
|
Abstract
Flaviviruses are a genus of (+)ssRNA (positive ssRNA) enveloped viruses that replicate in the cytoplasm of cells of diverse species from arthropods to mammals. Many are important human pathogens such as DENV-1-4 (dengue virus types 1-4), WNV (West Nile virus), YFV (yellow fever virus), JEV (Japanese encephalitis virus) and TBEV (tick-borne encephalitis). Given their RNA genomes it is not surprising that flaviviral life cycles revolve around critical RNA transactions. It is these we highlight in the present article. First, we summarize the mechanisms governing flaviviral replication and the central role of conserved RNA elements and viral protein-RNA interactions in RNA synthesis, translation and packaging. Secondly, we focus on how host RNA-binding proteins both benefit and inhibit flaviviral replication at different stages of their life cycle in mammalian hosts. Thirdly, we cover recent studies on viral non-coding RNAs produced in flavivirus-infected cells and how these RNAs affect various aspects of cellular RNA metabolism. Together, the article puts into perspective the central role of flaviviral RNAs in modulating both viral and cellular functions.
Collapse
|
45
|
Abstract
Dengue virus (DENV) is an emerging mosquito-borne human pathogen that affects millions of individuals each year by causing severe and potentially fatal syndromes. Despite intense research efforts, no approved vaccine or antiviral therapy is yet available. Overcoming this limitation requires detailed understanding of the intimate relationship between the virus and its host cell, providing the basis to devise optimal prophylactic and therapeutic treatment options. With the advent of novel high-throughput technologies including functional genomics, transcriptomics, proteomics, and lipidomics, new important insights into the DENV replication cycle and the interaction of this virus with its host cell have been obtained. In this chapter, we provide a comprehensive overview on the current status of the DENV research field, covering every step of the viral replication cycle with a particular focus on virus-host cell interaction. We will also review specific chemical inhibitors targeting cellular factors and processes of relevance for the DENV replication cycle and their possible exploitation for the development of next generation antivirals.
Collapse
|
46
|
Roby JA, Bielefeldt-Ohmann H, Prow NA, Chang DC, Hall RA, Khromykh AA. Increased expression of capsid protein in trans enhances production of single-round infectious particles by West Nile virus DNA vaccine candidate. J Gen Virol 2014; 95:2176-2191. [PMID: 24958626 DOI: 10.1099/vir.0.064121-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
West Nile virus (WNV; genus Flavivirus, family Flaviviridae) is an emerging pathogenic arbovirus responsible for outbreaks of encephalitis around the world. Whilst no vaccines are currently available to prevent WNV infection of humans, the use of cDNA copies of flavivirus RNA genomes with large internal deletions within the capsid (C) appears promising. C-deleted vaccines are able to replicate and secrete large amounts of non-infectious immunogenic subviral particles (SVPs) from transfected cells. We have previously generated a WNV DNA vaccine candidate pKUNdC/C where C-deleted WNV cDNA was placed under the control of one copy of the cytomegalovirus (CMV) promoter and the C gene was placed under the control of a second copy of the CMV promoter in the same plasmid DNA. This DNA was shown to generate single-round infectious particles (SRIPs) capable of delivering self-replicating C-deleted RNA producing SVPs to surrounding cells, thus enhancing the vaccine potential. However, the amounts of both SRIPs and SVPs produced from pKUNdC/C DNA were relatively low. In this investigation, we aimed at increasing SRIP production by optimizing trans-C expression via incorporating different forms of C and the use of a more powerful promoter. The construct containing an elongation factor EF1α promoter encoding an extended form of C was demonstrated to produce the highest titres of SRIPs and was immunogenic in mice. Additionally, SRIP and SVP titres were further improved via incorporation of a glycosylation motif in the envelope protein. The optimized DNA yielded ~100-fold greater titres of SRIPs than the original construct, thus providing a promising candidate for further vaccine evaluation.
Collapse
Affiliation(s)
- Justin A Roby
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia.,Australian Infectious Diseases Research Centre, University of Queensland, Australia
| | - Helle Bielefeldt-Ohmann
- School of Veterinary Sciences, University of Queensland, Australia.,Australian Infectious Diseases Research Centre, University of Queensland, Australia
| | - Natalie A Prow
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia.,Australian Infectious Diseases Research Centre, University of Queensland, Australia
| | - David C Chang
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia
| | - Roy A Hall
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia.,Australian Infectious Diseases Research Centre, University of Queensland, Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia.,Australian Infectious Diseases Research Centre, University of Queensland, Australia
| |
Collapse
|
47
|
Nazmi A, Dutta K, Hazra B, Basu A. Role of pattern recognition receptors in flavivirus infections. Virus Res 2014; 185:32-40. [PMID: 24657789 DOI: 10.1016/j.virusres.2014.03.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/11/2014] [Accepted: 03/11/2014] [Indexed: 12/24/2022]
Abstract
The flaviviral encephalitis has now become a major health concern in global scale. The efficient detection of viral infection and induction of the innate antiviral response by host's innate immune system are crucial to determine the outcome of infection. The intracellular pattern recognition receptors TLRs, RLRs, NLRs and CLRs play a central role in detection and initiation of robust antiviral response against flaviviral infection. Both cytoplasmic RLRs, RIG-I and MDA5 have been shown to be implicated in sensing flaviviral genomic RNA. Similarly among TLRs mainly TLR3 and TLR7 are known to respond in flaviviral infections as they are known to sense dsRNA and ssRNA moiety as their natural cognate ligand. Several studies have also shown the roles of NLRs and CLRs in mounting an innate antiviral response against flavivirus but, it is yet to be completely understood. Until now only few reports have implicated NLRs and CLRs in induction of antiviral and proinflammatory state following flaviviral infection. The current review therefore aims to comprehensively analyze past as well as current understanding on the role of PRRs in flaviviral infections.
Collapse
Affiliation(s)
- Arshed Nazmi
- National Brain Research Centre, Manesar, Haryana 122051, India.
| | - Kallol Dutta
- National Brain Research Centre, Manesar, Haryana 122051, India
| | - Bibhabasu Hazra
- National Brain Research Centre, Manesar, Haryana 122051, India
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana 122051, India.
| |
Collapse
|
48
|
Morrison J, García-Sastre A. STAT2 signaling and dengue virus infection. JAKSTAT 2014; 3:e27715. [PMID: 24778924 PMCID: PMC3995738 DOI: 10.4161/jkst.27715] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 12/24/2013] [Accepted: 01/03/2014] [Indexed: 12/28/2022] Open
Abstract
Dengue virus (DENV) is an important human pathogen whose byzantine relationship with the immune response is poorly understood. DENV causes dengue fever and dengue hemorrhagic fever/dengue shock syndrome, diseases for which palliative care is the only treatment. DENV immunopathogenesis studies are complicated by the lack of an immunocompetent small-animal model, and this has hindered anti-DENV drug and vaccine development. This review describes strategies that DENV uses to evade the type I interferon response and focuses on how data gained from the study of DENV NS5-mediated STAT2 degradation may be used to create immunocompetent DENV mouse models and design anti-DENV therapeutics.
Collapse
Affiliation(s)
- Juliet Morrison
- Department of Microbiology; University of Washington; Seattle, WA USA
| | - Adolfo García-Sastre
- Department of Microbiology; Icahn School of Medicine at Mount Sinai; New York, NY USA ; Global Health and Emerging Pathogens Institute; Icahn School of Medicine at Mount Sinai; New York, NY USA ; Department of Medicine; Icahn School of Medicine at Mount Sinai; New York, NY USA
| |
Collapse
|
49
|
Zinzula L, Tramontano E. Strategies of highly pathogenic RNA viruses to block dsRNA detection by RIG-I-like receptors: hide, mask, hit. Antiviral Res 2013; 100:615-35. [PMID: 24129118 PMCID: PMC7113674 DOI: 10.1016/j.antiviral.2013.10.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/24/2013] [Accepted: 10/04/2013] [Indexed: 12/24/2022]
Abstract
dsRNA species are byproducts of RNA virus replication and/or transcription. Prompt detection of dsRNA by RIG-I like receptors (RLRs) is a hallmark of the innate immune response. RLRs activation triggers production of the type I interferon (IFN)-based antiviral response. Highly pathogenic RNA viruses encode proteins that block the RLRs pathway. Hide, mask and hit are 3 strategies of RNA viruses to avoid immune system activation.
Double-stranded RNA (dsRNA) is synthesized during the course of infection by RNA viruses as a byproduct of replication and transcription and acts as a potent trigger of the host innate antiviral response. In the cytoplasm of the infected cell, recognition of the presence of viral dsRNA as a signature of “non-self” nucleic acid is carried out by RIG-I-like receptors (RLRs), a set of dedicated helicases whose activation leads to the production of type I interferon α/β (IFN-α/β). To overcome the innate antiviral response, RNA viruses encode suppressors of IFN-α/β induction, which block RLRs recognition of dsRNA by means of different mechanisms that can be categorized into: (i) dsRNA binding and/or shielding (“hide”), (ii) dsRNA termini processing (“mask”) and (iii) direct interaction with components of the RLRs pathway (“hit”). In light of recent functional, biochemical and structural findings, we review the inhibition mechanisms of RLRs recognition of dsRNA displayed by a number of highly pathogenic RNA viruses with different disease phenotypes such as haemorrhagic fever (Ebola, Marburg, Lassa fever, Lujo, Machupo, Junin, Guanarito, Crimean-Congo, Rift Valley fever, dengue), severe respiratory disease (influenza, SARS, Hendra, Hantaan, Sin Nombre, Andes) and encephalitis (Nipah, West Nile).
Collapse
Affiliation(s)
- Luca Zinzula
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella di Monserrato, SS554, 09042 Monserrato (Cagliari), Italy.
| | | |
Collapse
|
50
|
Abraham R, Mudaliar P, Padmanabhan A, Sreekumar E. Induction of cytopathogenicity in human glioblastoma cells by chikungunya virus. PLoS One 2013; 8:e75854. [PMID: 24086645 PMCID: PMC3783433 DOI: 10.1371/journal.pone.0075854] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 08/22/2013] [Indexed: 01/01/2023] Open
Abstract
Chikungunya virus (CHIKV), an arthritogenic old-world alphavirus, has been implicated in the central nervous system (CNS) infection in infants and elderly patients. Astrocytes are the major immune cells of the brain parenchyma that mediate inflammation. In the present study we found that a local isolate of CHIKV infect and activate U-87 MG cells, a glioblastoma cell line of human astrocyte origin. The infection kinetics were similar in infected U-87 MG cells and the human embryo kidney (HEK293) cells as indicated by immunofluorescence and plaque assays, 24h post-infection (p.i.). In infected U-87 MG cells, apoptosis was detectable from 48h p.i. evidenced by DNA fragmentation, PARP cleavage, loss of mitochondrial membrane potential, nuclear condensation and visible cytopathic effects in a dose and time-dependent manner. XBP1 mRNA splicing and eIF2α phosphorylation studies indicated the occurrence of endoplasmic reticulum stress in infected cells. In U-87 MG cells stably expressing a green fluorescent protein-tagged light chain-3 (GFP-LC3) protein, CHIKV infection showed increased autophagy response. The infection led to an enhanced expression of the mRNA transcripts of the pro-inflammatory cytokines IL-1β, TNF-α, IL-6 and CXCL9 within 24h p.i. Significant up-regulation of the proteins of RIG-I like receptor (RLR) pathway, such as RIG-I and TRAF-6, was observed indicating the activation of the cytoplasmic-cellular innate immune response. The overall results show that the U-87 MG cell line is a potential in vitro model for in depth study of these molecular pathways in response to CHIKV infection. The responses in these cells of CNS origin, which are inherently defective in Type I interferon response, could be analogous to that occurring in infants and very old patients who also have a compromised interferon-response. The results also point to the intriguing possibility of using this virus for studies to develop oncolytic virus therapy approaches against glioblastoma, a highly aggressive malignancy.
Collapse
Affiliation(s)
- Rachy Abraham
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Prashant Mudaliar
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Aiswaria Padmanabhan
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Easwaran Sreekumar
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| |
Collapse
|