101
|
Abstract
Filovirus small animal disease models have so far been developed in laboratory mice, guinea pigs, and hamsters. Since immunocompetent rodents do not exhibit overt signs of disease following infection with wild-type filoviruses isolated from humans, rodent models have been established using adapted viruses produced through sequential passage in rodents. Rodent-adapted viruses target the same cells/tissues as the wild-type viruses, making rodents invaluable basic research tools for studying filovirus pathogenesis. Moreover, comparative analyses using wild-type and rodent-adapted viruses have provided beneficial insights into the molecular mechanisms of pathogenicity and acquisition of species-specific virulence. Additionally, wild-type filovirus infections in immunodeficient rodents have provided a better understanding of the host factors required for resistance to filovirus infection and of the immune response against the infection. This chapter provides comprehensive information on the filovirus rodent models and rodent-adapted filoviruses. Specifically, we summarize the clinical and pathological features of filovirus infections in all rodent models described to date, including the recently developed humanized and collaborative cross (CC) resource recombinant inbred (RI) intercrossed (CC-RIX) mouse models. We also cover the molecular determinants responsible for adaptation and virulence acquisition in a number of rodent-adapted filoviruses. This chapter clearly defines the characteristic and advantages/disadvantages of rodent models, helping to evaluate the practical use of rodent models in future filovirus studies.
Collapse
|
102
|
Pleet ML, DeMarino C, Stonier SW, Dye JM, Jacobson S, Aman MJ, Kashanchi F. Extracellular Vesicles and Ebola Virus: A New Mechanism of Immune Evasion. Viruses 2019; 11:v11050410. [PMID: 31052499 PMCID: PMC6563240 DOI: 10.3390/v11050410] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023] Open
Abstract
Ebola virus (EBOV) disease can result in a range of symptoms anywhere from virtually asymptomatic to severe hemorrhagic fever during acute infection. Additionally, spans of asymptomatic persistence in recovering survivors is possible, during which transmission of the virus may occur. In acute infection, substantial cytokine storm and bystander lymphocyte apoptosis take place, resulting in uncontrolled, systemic inflammation in affected individuals. Recently, studies have demonstrated the presence of EBOV proteins VP40, glycoprotein (GP), and nucleoprotein (NP) packaged into extracellular vesicles (EVs) during infection. EVs containing EBOV proteins have been shown to induce apoptosis in recipient immune cells, as well as contain pro-inflammatory cytokines. In this manuscript, we review the current field of knowledge on EBOV EVs including the mechanisms of their biogenesis, their cargo and their effects in recipient cells. Furthermore, we discuss some of the effects that may be induced by EBOV EVs that have not yet been characterized and highlight the remaining questions and future directions.
Collapse
Affiliation(s)
- Michelle L Pleet
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA.
| | - Catherine DeMarino
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA.
| | - Spencer W Stonier
- Department, Emergent BioSolutions, Gaithersburg, MD 20879, USA.
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA.
| | - John M Dye
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA.
| | - Steven Jacobson
- Viral Immunology Section, Neuroimmunology Branch, National Institute for Neurological Disease and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - M Javad Aman
- Department. Integrated BioTherapeutics, Inc., Gaithersburg, MD 20850, USA.
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA.
| |
Collapse
|
103
|
Edwards MR, Basler CF. Current status of small molecule drug development for Ebola virus and other filoviruses. Curr Opin Virol 2019; 35:42-56. [PMID: 31003196 PMCID: PMC6556423 DOI: 10.1016/j.coviro.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
The filovirus family includes some of the deadliest viruses known, including Ebola virus and Marburg virus. These viruses cause periodic outbreaks of severe disease that can be spread from person to person, making the filoviruses important public health threats. There remains a need for approved drugs that target all or most members of this virus family. Small molecule inhibitors that target conserved functions hold promise as pan-filovirus therapeutics. To date, compounds that effectively target virus entry, genome replication, gene expression, and virus egress have been described. The most advanced inhibitors are nucleoside analogs that target viral RNA synthesis reactions.
Collapse
Affiliation(s)
- Megan R Edwards
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States.
| |
Collapse
|
104
|
Rahim MN, Wang M, Wang T, He S, Griffin BD, Kobasa D, Yang R, Du Z, Qiu X. Generation and Characterization of Anti-Filovirus Nucleoprotein Monoclonal Antibodies. Viruses 2019; 11:v11030259. [PMID: 30875741 PMCID: PMC6466340 DOI: 10.3390/v11030259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/03/2019] [Accepted: 03/08/2019] [Indexed: 12/27/2022] Open
Abstract
Filoviruses cause lethal hemorrhagic fever in humans. The filovirus nucleoprotein (NP) is expressed in high abundance in infected cells and is essential for virus replication. To generate anti-filovirus monoclonal antibodies (mAbs) against the NP, mice were immunized with peptides known as B-cell epitopes corresponding to different filovirus NPs, and hybridomas were screened using FLAG-tagged filovirus NP constructs. Numerous mAbs were identified, isotyped, and characterized. The anti-NP mAbs demonstrated different ranges of binding affinities to various filovirus NPs. Most of the clones specifically detected both recombinant and wild-type NPs from different filoviruses, including Ebola (EBOV), Sudan (SUDV), Bundibugyo (BDBV), Marburg (MARV), Tai Forest (TAFV), and Reston (RESTV) viruses in western blot analysis. The mAbs were also able to detect native NPs within the cytoplasm of infected cells by immunofluorescence confocal microscopy. Thus, this panel of mAbs represents an important set of tools that may be potentially useful for diagnosing filovirus infection, characterizing virus replication, and detecting NP–host protein interactions.
Collapse
Affiliation(s)
- Md Niaz Rahim
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada.
- Department of Medical Microbiology, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| | - Min Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Tong Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada.
| | - Bryan D Griffin
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada.
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada.
- Department of Medical Microbiology, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Zongmin Du
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB, R3E 3R2, Canada.
- Department of Medical Microbiology, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| |
Collapse
|
105
|
Role of Type I Interferons on Filovirus Pathogenesis. Vaccines (Basel) 2019; 7:vaccines7010022. [PMID: 30791589 PMCID: PMC6466283 DOI: 10.3390/vaccines7010022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/15/2019] [Indexed: 01/19/2023] Open
Abstract
Filoviruses, such as Ebola and Marburg virus, encode viral proteins with the ability to counteract the type I interferon (IFN-I) response. These IFN-I antagonist proteins are crucial to ensure virus replication, prevent an antiviral state in infected and bystander cells, and impair the ability of antigen-presenting cells to initiate adaptive immune responses. However, in recent years, a number of studies have underscored the conflicting data between in vitro studies and in vivo data obtained in animal models and clinical studies during outbreaks. This review aims to summarize these data and to discuss the relative contributions of IFN-α and IFN-β to filovirus pathogenesis in animal models and humans. Finally, we evaluate the putative utilization of IFN-I in post-exposure therapy and its implications as a biomarker of vaccine efficacy.
Collapse
|
106
|
Shipley R, Wright E, Selden D, Wu G, Aegerter J, Fooks AR, Banyard AC. Bats and Viruses: Emergence of Novel Lyssaviruses and Association of Bats with Viral Zoonoses in the EU. Trop Med Infect Dis 2019; 4:tropicalmed4010031. [PMID: 30736432 PMCID: PMC6473451 DOI: 10.3390/tropicalmed4010031] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/25/2022] Open
Abstract
Bats in the EU have been associated with several zoonotic viral pathogens of significance to both human and animal health. Virus discovery continues to expand the existing understating of virus classification, and the increased interest in bats globally as reservoirs or carriers of zoonotic agents has fuelled the continued detection and characterisation of new lyssaviruses and other viral zoonoses. Although the transmission of lyssaviruses from bat species to humans or terrestrial species appears rare, interest in these viruses remains, through their ability to cause the invariably fatal encephalitis—rabies. The association of bats with other viral zoonoses is also of great interest. Much of the EU is free of terrestrial rabies, but several bat species harbor lyssaviruses that remain a risk to human and animal health. Whilst the rabies virus is the main cause of rabies globally, novel related viruses continue to be discovered, predominantly in bat populations, that are of interest purely through their classification within the lyssavirus genus alongside the rabies virus. Although the rabies virus is principally transmitted from the bite of infected dogs, these related lyssaviruses are primarily transmitted to humans and terrestrial carnivores by bats. Even though reports of zoonotic viruses from bats within the EU are rare, to protect human and animal health, it is important characterise novel bat viruses for several reasons, namely: (i) to investigate the mechanisms for the maintenance, potential routes of transmission, and resulting clinical signs, if any, in their natural hosts; (ii) to investigate the ability of existing vaccines, where available, to protect against these viruses; (iii) to evaluate the potential for spill over and onward transmission of viral pathogens in novel terrestrial hosts. This review is an update on the current situation regarding zoonotic virus discovery within bats in the EU, and provides details of potential future mechanisms to control the threat from these deadly pathogens.
Collapse
Affiliation(s)
- Rebecca Shipley
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), KT15 3NB Weybridge-London, UK.
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG Brighton, UK.
| | - Edward Wright
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG Brighton, UK.
| | - David Selden
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), KT15 3NB Weybridge-London, UK.
| | - Guanghui Wu
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), KT15 3NB Weybridge-London, UK.
| | - James Aegerter
- APHA - National Wildlife Management Centre, Wildlife Epidemiology and Modelling, Sand Hutton, YO41 1LZ York, UK.
| | - Anthony R Fooks
- Institute for Infection and Immunity, St. George's Hospital Medical School, University of London, London, SW17 0RE, UK.
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK.
| | - Ashley C Banyard
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), KT15 3NB Weybridge-London, UK.
- School of Life Sciences, University of Sussex, Falmer, BN1 9QG Brighton, UK.
- Institute for Infection and Immunity, St. George's Hospital Medical School, University of London, London, SW17 0RE, UK.
| |
Collapse
|
107
|
Abstract
Ebolaviruses have gained much attention recently due to the outbreak from 2014 through 2016. The related marburgviruses also have been responsible for large outbreaks with high case fatality rates. The purpose of this article is to provide the clinical laboratory scientist with a review of the most current developments in marburgvirus research. The PubMed database was reviewed using the keywords "Marburg virus," "Ravn virus," and "marburgviruses," with publication dates from January 1, 2015 through June 20, 2017. The search yielded 345 articles. In total, 52 articles met the inclusion criteria and were reviewed. Advances have been made in the areas of ecology and host reservoir studies, seroprevalence studies, pathology and pathogenesis studies, laboratory assay development, and treatment and vaccine development. Marburgviruses are highly lethal viruses that pose a significant threat to the human population. Although numerous advances have been made, there are still large gaps in knowledge, and it is imperative that scientists gain more information to fully understand virus/host interactions. An approved vaccine and treatment remain elusive.
Collapse
|
108
|
Chanthamontri CK, Jordan DS, Wang W, Wu C, Lin Y, Brett TJ, Gross ML, Leung DW. The Ebola Viral Protein 35 N-Terminus Is a Parallel Tetramer. Biochemistry 2019; 58:657-664. [PMID: 30592210 DOI: 10.1021/acs.biochem.8b01154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Members of Mononegavirales, the order that includes nonsegmented negative sense RNA viruses (NNSVs), encode a small number of multifunctional proteins. In members of the Filoviridae family, virus protein 35 (VP35) facilitates immune evasion and functions as an obligatory cofactor for viral RNA synthesis. VP35 functions in a manner orthologous to that of phosphoproteins from other NNSVs. Although the critical roles of Ebola viral VP35 (eVP35) in immune evasion and RNA synthesis are well-appreciated, a complete understanding of its organization and its role in carrying out its many functions has yet to be fully realized. In particular, we currently lack information about the role of the oligomerization domain within eVP35. To address this limitation, we report here an investigation of the oligomer structure of eVP35 using hybrid methods that include multiangle light scattering, small-angle X-ray scattering, and cross-linking coupled with mass spectrometry to determine the shape and orientation of the eVP35 oligomer. Our integrative results are consistent with a parallel tetramer in which the N-terminal regions that are required for RNA synthesis are all oriented in the same direction. Furthermore, these results define a framework for targeting the symmetric tetramer for structure-based antiviral discovery.
Collapse
Affiliation(s)
- Chamnongsak Ken Chanthamontri
- Department of Chemistry , Washington University in St. Louis , Box 1134, One Brookings Drive , St. Louis , Missouri 63130 , United States
| | - David S Jordan
- Department of Pathology and Immunology , Washington University School of Medicine in St. Louis , St. Louis , Missouri 63110 , United States
| | - Wenjie Wang
- Department of Pathology and Immunology , Washington University School of Medicine in St. Louis , St. Louis , Missouri 63110 , United States
| | - Chao Wu
- Department of Pathology and Immunology , Washington University School of Medicine in St. Louis , St. Louis , Missouri 63110 , United States
| | - Yanchun Lin
- Department of Chemistry , Washington University in St. Louis , Box 1134, One Brookings Drive , St. Louis , Missouri 63130 , United States
| | - Tom J Brett
- Department of Medicine , Washington University School of Medicine in St. Louis , St. Louis , Missouri 63110 , United States
| | - Michael L Gross
- Department of Chemistry , Washington University in St. Louis , Box 1134, One Brookings Drive , St. Louis , Missouri 63130 , United States
| | - Daisy W Leung
- Department of Pathology and Immunology , Washington University School of Medicine in St. Louis , St. Louis , Missouri 63110 , United States
| |
Collapse
|
109
|
Reynard S, Journeaux A, Gloaguen E, Schaeffer J, Varet H, Pietrosemoli N, Mateo M, Baillet N, Laouenan C, Raoul H, Mullaert J, Baize S. Immune parameters and outcomes during Ebola virus disease. JCI Insight 2019; 4:125106. [PMID: 30626757 DOI: 10.1172/jci.insight.125106] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/29/2018] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The West African Ebola virus epidemic from 2014-2016 highlighted the lack of knowledge about the pathogenicity of the virus and the factors responsible for outcome. A performant and rapid diagnosis is of crucial importance, as is overcoming the difficulty of providing high-quality patient management during such an extensive outbreak. Here, we propose to study the role of the immune mediators during Ebola virus disease and to define some molecules of importance in the outcome. METHODS Plasma from Guinean patients sampled during the outbreak were analyzed using RT-qPCR, magnetic bead assay, ELISA, and high-quality statistical analyses. We also performed a transcriptomic analysis in leukocytes samples. Therefore, we deeply characterized the immune responses involved in Ebola virus disease. RESULTS We evaluated the immune patterns depending on the outcome of the disease. Survivors presented an efficient and well-balanced immune response, whereas fatalities were characterized by an intense inflammatory response, overexpression of multiple cytokines, and a "chemokine storm." The plasma concentration of most of the parameters tested increased until death. Statistical analyses also allowed us to define a panel of markers highly predictive of outcome. CONCLUSION The immune response observed in fatalities was highly similar to that characterizing septic shock syndrome. Our results suggest that immune responses can play a major pathogenic role during severe Ebola virus infection and argue in favor of therapeutic approaches that act on both viral replication and the induction of shock syndrome. FUNDING French Ministry of Foreign Affairs, the Agence Française de Développement, and the Institut Pasteur.
Collapse
Affiliation(s)
- Stéphanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Alexandra Journeaux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Emilie Gloaguen
- Infection Antimicrobials Modelling Evolution, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Justine Schaeffer
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Hugo Varet
- Institut Pasteur, Hub Bioinformatique et Biostatistique, Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI, USR 3756 IP CNRS, Paris, France
| | - Natalia Pietrosemoli
- Institut Pasteur, Hub Bioinformatique et Biostatistique, Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI, USR 3756 IP CNRS, Paris, France
| | - Mathieu Mateo
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Nicolas Baillet
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| | - Cédric Laouenan
- Infection Antimicrobials Modelling Evolution, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Bichat Claude Bernard, Paris, France
| | - Hervé Raoul
- Laboratoire P4 Jean Mérieux-INSERM, INSERM, Lyon, France
| | - Jimmy Mullaert
- Infection Antimicrobials Modelling Evolution, UMR 1137, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Centre International de Recherche en Infectiologie, Université Lyon I, INSERM, CNRS, ENS Lyon, Lyon, France
| |
Collapse
|
110
|
Yang X, Wang X, Song Y, Zhou P, Li D, Zhang C, Jin X, Huang Z, Zhou D. Chimpanzee adenoviral vector prime-boost regimen elicits potent immune responses against Ebola virus in mice and rhesus macaques. Emerg Microbes Infect 2019; 8:1086-1097. [PMID: 31339465 PMCID: PMC6711196 DOI: 10.1080/22221751.2019.1644968] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 07/11/2019] [Indexed: 12/19/2022]
Abstract
In the last few decades, Ebola virus (EBOV) has emerged periodically and infected people in Africa, resulting in an extremely high mortality rate. With no available prophylaxis or cure so far, a highly effective Ebola vaccine is urgently needed. In this study, we developed a novel chimpanzee adenovirus-based prime-boost vaccine by exploiting two recombinant replication-deficient chimpanzee adenoviral vectors, AdC7 and AdC68, which express glycoproteins (GP) of the EBOV strain identified in the 2014 outbreak. Our results indicated that a single immunization using AdC7 or AdC68 could stimulate potent EBOV-specific antibody responses, whereas the AdC7 prime-AdC68 boost regimen induced much stronger and sustained humoral and cellular immune responses in both mice and rhesus monkeys, compared with AdC7 or AdC68 single vaccination or the AdC68 prime-AdC7 boost regimen. This prime-boost vaccine could also protect mice from the simulated infection with EBOV-like particle (EBOVLP) in biosafety level 2 (BSL-2) laboratories, and antibodies from the prime-boost immunized rhesus macaques could passively provide protection against EBOVLP infection. Altogether, our results show that the AdC7 prime-AdC68 boost vaccine is a promising candidate for further development to combat EBOV infections.
Collapse
Affiliation(s)
- Xi Yang
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Xiang Wang
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Yufeng Song
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Ping Zhou
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Dapeng Li
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Chao Zhang
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Xia Jin
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Zhong Huang
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Dongming Zhou
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
111
|
Lindstrom A, Anantpadma M, Baker L, Raghavendra NM, Davey R, Davisson VJ. Phenotypic Prioritization of Diphyllin Derivatives That Block Filoviral Cell Entry by Vacuolar (H + )-ATPase Inhibition. ChemMedChem 2018; 13:2664-2676. [PMID: 30335906 PMCID: PMC6387451 DOI: 10.1002/cmdc.201800587] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Indexed: 01/28/2023]
Abstract
Many viruses use endosomal pathways to gain entry into cells and propagate infection. Sensing of endosomal acidification is a trigger for the release of many virus cores into the cell cytosol. Previous efforts with inhibitors of vacuolar ATPase have been shown to block endosomal acidification and affect viral entry, albeit with limited potential for therapeutic selectivity. In this study, four novel series of derivatives of the vacuolar ATPase inhibitor diphyllin were synthesized to assess their potential for enhancing potency and anti-filoviral activity over cytotoxicity. Derivatives that suitably blocked cellular entry of Ebola pseudotyped virus were further evaluated as inhibitors of endosomal acidification and isolated human vacuolar ATPase activity. Several compounds with significant increases in potency over diphyllin in these assays also separated from cytotoxic doses in human cell models by >100-fold. Finally, three derivatives were shown to be inhibitors of replication-competent Ebola viral entry into primary macrophages with similar potencies and enhanced selectivity toward antiviral activity.
Collapse
Affiliation(s)
- Aaron Lindstrom
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Manu Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, 8715 West Military Drive, San Antonio, TX, 78227, USA
- Current address: Department of Microbiology, Boston University, Boston, MA, 02118, USA
| | - Logan Baker
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - N M Raghavendra
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Robert Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, 8715 West Military Drive, San Antonio, TX, 78227, USA
- Current address: Department of Microbiology, Boston University, Boston, MA, 02118, USA
| | - Vincent Jo Davisson
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
112
|
Batra J, Hultquist JF, Liu D, Shtanko O, Von Dollen J, Satkamp L, Jang GM, Luthra P, Schwarz TM, Small GI, Arnett E, Anantpadma M, Reyes A, Leung DW, Kaake R, Haas P, Schmidt CB, Schlesinger LS, LaCount DJ, Davey RA, Amarasinghe GK, Basler CF, Krogan NJ. Protein Interaction Mapping Identifies RBBP6 as a Negative Regulator of Ebola Virus Replication. Cell 2018; 175:1917-1930.e13. [PMID: 30550789 PMCID: PMC6366944 DOI: 10.1016/j.cell.2018.08.044] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/17/2018] [Accepted: 08/17/2018] [Indexed: 01/21/2023]
Abstract
Ebola virus (EBOV) infection often results in fatal illness in humans, yet little is known about how EBOV usurps host pathways during infection. To address this, we used affinity tag-purification mass spectrometry (AP-MS) to generate an EBOV-host protein-protein interaction (PPI) map. We uncovered 194 high-confidence EBOV-human PPIs, including one between the viral transcription regulator VP30 and the host ubiquitin ligase RBBP6. Domain mapping identified a 23 amino acid region within RBBP6 that binds to VP30. A crystal structure of the VP30-RBBP6 peptide complex revealed that RBBP6 mimics the viral nucleoprotein (NP) binding to the same interface of VP30. Knockdown of endogenous RBBP6 stimulated viral transcription and increased EBOV replication, whereas overexpression of either RBBP6 or the peptide strongly inhibited both. These results demonstrate the therapeutic potential of biologics that target this interface and identify additional PPIs that may be leveraged for novel therapeutic strategies.
Collapse
Affiliation(s)
- Jyoti Batra
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Judd F Hultquist
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA; Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dandan Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63105, USA
| | - Olena Shtanko
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - John Von Dollen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Laura Satkamp
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Gwendolyn M Jang
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Priya Luthra
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Toni M Schwarz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gabriel I Small
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63105, USA
| | - Eusondia Arnett
- Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Manu Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Ann Reyes
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63105, USA
| | - Robyn Kaake
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Paige Haas
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Carson B Schmidt
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA
| | | | - Douglas J LaCount
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Robert A Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63105, USA
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; J. David Gladstone Institutes, San Francisco, CA 94158, USA.
| |
Collapse
|
113
|
Basler CF, Krogan NJ, Leung DW, Amarasinghe GK. Virus and host interactions critical for filoviral RNA synthesis as therapeutic targets. Antiviral Res 2018; 162:90-100. [PMID: 30550800 DOI: 10.1016/j.antiviral.2018.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/05/2018] [Accepted: 12/08/2018] [Indexed: 01/24/2023]
Abstract
Filoviruses, which include Ebola virus (EBOV) and Marburg virus, are negative-sense RNA viruses associated with sporadic outbreaks of severe viral hemorrhagic fever characterized by uncontrolled virus replication. The extreme virulence and emerging nature of these zoonotic pathogens make them a significant threat to human health. Replication of the filovirus genome and production of viral RNAs require the function of a complex of four viral proteins, the nucleoprotein (NP), viral protein 35 (VP35), viral protein 30 (VP30) and large protein (L). The latter performs the enzymatic activities required for production of viral RNAs and capping of viral mRNAs. Although it has been recognized that interactions between the virus-encoded components of the EBOV RNA polymerase complex are required for viral RNA synthesis reactions, specific molecular details have, until recently, been lacking. New efforts have combined structural biology and molecular virology to reveal in great detail the molecular basis for critical protein-protein interactions (PPIs) necessary for viral RNA synthesis. These efforts include recent studies that have identified a range of interacting host factors and in some instances demonstrated unique mechanisms by which they act. For a select number of these interactions, combined use of mutagenesis, over-expressing of peptides corresponding to PPI interfaces and identification of small molecules that disrupt PPIs have demonstrated the functional significance of virus-virus and virus-host PPIs and suggest several as potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| | - Nevan J Krogan
- Quantitative Biosciences Institute (QBI), UCSF, San Francisco, CA, USA; Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
114
|
Schindell BG, Webb AL, Kindrachuk J. Persistence and Sexual Transmission of Filoviruses. Viruses 2018; 10:E683. [PMID: 30513823 PMCID: PMC6316729 DOI: 10.3390/v10120683] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022] Open
Abstract
There is an increasing frequency of reports regarding the persistence of the Ebola virus (EBOV) in Ebola virus disease (EVD) survivors. During the 2014⁻2016 West African EVD epidemic, sporadic transmission events resulted in the initiation of new chains of human-to-human transmission. Multiple reports strongly suggest that these re-emergences were linked to persistent EBOV infections and included sexual transmission from EVD survivors. Asymptomatic infection and long-term viral persistence in EVD survivors could result in incidental introductions of the Ebola virus in new geographic regions and raise important national and local public health concerns. Alarmingly, although the persistence of filoviruses and their potential for sexual transmission have been documented since the emergence of such viruses in 1967, there is limited knowledge regarding the events that result in filovirus transmission to, and persistence within, the male reproductive tract. Asymptomatic infection and long-term viral persistence in male EVD survivors could lead to incidental transfer of EBOV to new geographic regions, thereby generating widespread outbreaks that constitute a significant threat to national and global public health. Here, we review filovirus testicular persistence and discuss the current state of knowledge regarding the rates of persistence in male survivors, and mechanisms underlying reproductive tract localization and sexual transmission.
Collapse
Affiliation(s)
- Brayden G Schindell
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Andrew L Webb
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Jason Kindrachuk
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
115
|
Haddock E, Feldmann H, Marzi A. Ebola Virus Infection in Commonly Used Laboratory Mouse Strains. J Infect Dis 2018; 218:S453-S457. [PMID: 29878128 PMCID: PMC6249562 DOI: 10.1093/infdis/jiy208] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mouse model for Ebola virus (EBOV) is an established and often used animal model for countermeasure development. Although it has its limitations, it recapitulates certain key features of human EBOV disease and principally shows uniform lethality. However, in the recent past, several studies reported surviving animals when evaluating treatment or vaccine approaches. Therefore, we analyzed the severity of disease and lethality of mouse-adapted (MA-) EBOV infection in 6 different mouse strains. We identified outbred CD-1 mice to be the only strain tested resulting in uniform lethality when infected with different doses of MA-EBOV or reverse genetics-generated MA-EBOV. In contrast, infection of different inbred mouse strains resulted in partial survival depending on virus and dose. Of these inbred strains, 129 mice provided the most consistent model. Our study provides a helpful dataset when planning EBOV mouse studies for countermeasure efficacy testing and highlights the limitations of certain mouse strains as EBOV models.
Collapse
Affiliation(s)
- Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| |
Collapse
|
116
|
Younan P, Iampietro M, Santos RI, Ramanathan P, Popov VL, Bukreyev A. Role of Transmembrane Protein 16F in the Incorporation of Phosphatidylserine Into Budding Ebola Virus Virions. J Infect Dis 2018; 218:S335-S345. [PMID: 30289531 PMCID: PMC6249587 DOI: 10.1093/infdis/jiy485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Viral apoptotic mimicry, which is defined by exposure of phosphatidylserine (PtdSer) into the outer leaflet of budding enveloped viruses, increases viral tropism, infectivity and promotes immune evasion. Here, we report that the calcium (Ca2+)-dependent scramblase, transmembrane protein 16F (TMEM16F), is responsible for the incorporation of PtdSer into virion membranes during Ebola virus infection. Infection of Huh7 cells with Ebola virus resulted in a pronounced increase in plasma membrane-associated PtdSer, which was demonstrated to be dependent on TMEM16F function. Analysis of virions using imaging flow cytometry revealed that short hairpin RNA-mediated down-regulation of TMEM16F function directly reduced virion-associated PtdSer. Taken together, these studies demonstrate that TMEM16F is a central cellular factor in the exposure of PtdSer in the outer leaflet of viral membranes.
Collapse
Affiliation(s)
- Patrick Younan
- Departments of Pathology, University of Texas Medical Branch, Galveston
- Departments of Galveston National Laboratory, University of Texas Medical Branch, Galveston
- Departments of University of Texas Medical Branch, Galveston
| | - Mathieu Iampietro
- Departments of Pathology, University of Texas Medical Branch, Galveston
- Departments of Galveston National Laboratory, University of Texas Medical Branch, Galveston
- Departments of University of Texas Medical Branch, Galveston
| | - Rodrigo I Santos
- Departments of Pathology, University of Texas Medical Branch, Galveston
- Departments of Galveston National Laboratory, University of Texas Medical Branch, Galveston
- Departments of University of Texas Medical Branch, Galveston
| | - Palaniappan Ramanathan
- Departments of Pathology, University of Texas Medical Branch, Galveston
- Departments of Galveston National Laboratory, University of Texas Medical Branch, Galveston
- Departments of University of Texas Medical Branch, Galveston
| | - Vsevolod L Popov
- Departments of Pathology, University of Texas Medical Branch, Galveston
- Departments of University of Texas Medical Branch, Galveston
| | - Alexander Bukreyev
- Departments of Pathology, University of Texas Medical Branch, Galveston
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston
- Departments of Galveston National Laboratory, University of Texas Medical Branch, Galveston
- Departments of University of Texas Medical Branch, Galveston
| |
Collapse
|
117
|
Pleet ML, Erickson J, DeMarino C, Barclay RA, Cowen M, Lepene B, Liang J, Kuhn JH, Prugar L, Stonier SW, Dye JM, Zhou W, Liotta LA, Aman MJ, Kashanchi F. Ebola Virus VP40 Modulates Cell Cycle and Biogenesis of Extracellular Vesicles. J Infect Dis 2018; 218:S365-S387. [PMID: 30169850 PMCID: PMC6249571 DOI: 10.1093/infdis/jiy472] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Ebola virus (EBOV) mainly targets myeloid cells; however, extensive death of T cells is often observed in lethal infections. We have previously shown that EBOV VP40 in exosomes causes recipient immune cell death. Methods Using VP40-producing clones, we analyzed donor cell cycle, extracellular vesicle (EV) biogenesis, and recipient immune cell death. Transcription of cyclin D1 and nuclear localization of VP40 were examined via kinase and chromatin immunoprecipitation assays. Extracellular vesicle contents were characterized by mass spectrometry, cytokine array, and western blot. Biosafety level-4 facilities were used for wild-type Ebola virus infection studies. Results VP40 EVs induced apoptosis in recipient T cells and monocytes. VP40 clones were accelerated in growth due to cyclin D1 upregulation, and nuclear VP40 was found bound to the cyclin D1 promoter. Accelerated cell cycling was related to EV biogenesis, resulting in fewer but larger EVs. VP40 EV contents were enriched in ribonucleic acid-binding proteins and cytokines (interleukin-15, transforming growth factor-β1, and interferon-γ). Finally, EBOV-infected cell and animal EVs contained VP40, nucleoprotein, and glycoprotein. Conclusions Nuclear VP40 upregulates cyclin D1 levels, resulting in dysregulated cell cycle and EV biogenesis. Packaging of cytokines and EBOV proteins into EVs from infected cells may be responsible for the decimation of immune cells during EBOV pathogenesis.
Collapse
Affiliation(s)
- Michelle L Pleet
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, Virginia
| | - James Erickson
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, Virginia
| | - Catherine DeMarino
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, Virginia
| | - Robert A Barclay
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, Virginia
| | - Maria Cowen
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, Virginia
| | | | - Janie Liang
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland
| | - Laura Prugar
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland
| | - Spencer W Stonier
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland
| | - John M Dye
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, Virginia
| | - M Javad Aman
- Integrated BioTherapeutics, Inc., Gaithersburg, Maryland
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, Virginia
| |
Collapse
|
118
|
Zinzula L, Nagy I, Orsini M, Weyher-Stingl E, Bracher A, Baumeister W. Structures of Ebola and Reston Virus VP35 Oligomerization Domains and Comparative Biophysical Characterization in All Ebolavirus Species. Structure 2018; 27:39-54.e6. [PMID: 30482729 DOI: 10.1016/j.str.2018.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/18/2018] [Accepted: 09/19/2018] [Indexed: 12/12/2022]
Abstract
The multifunctional virion protein 35 (VP35) of ebolaviruses is a critical determinant of virulence and pathogenesis indispensable for viral replication and host innate immune evasion. Essential for VP35 function is homo-oligomerization via a coiled-coil motif. Here we report crystal structures of VP35 oligomerization domains from the prototypic Ebola virus (EBOV) and the non-pathogenic Reston virus (RESTV), together with a comparative biophysical characterization of the domains from all known species of the Ebolavirus genus. EBOV and RESTV VP35 oligomerization domains form bipartite parallel helix bundles with a canonical coiled coil in the N-terminal half and increased plasticity in the highly conserved C-terminal half. The domain assembles into trimers and tetramers in EBOV, whereas it exclusively forms tetramers in all other ebolavirus species. Substitution of coiled-coil leucine residues critical for immune antagonism leads to aberrant oligomerization. A conserved arginine involved in inter-chain salt bridges stabilizes the VP35 oligomerization domain and modulates between coiled-coil oligomeric states.
Collapse
Affiliation(s)
- Luca Zinzula
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - István Nagy
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Massimiliano Orsini
- Istituto Zooprofilattico dell'Abruzzo e del Molise, Campo Boario, 64100 Teramo, Italy
| | - Elisabeth Weyher-Stingl
- The Max-Planck Institute of Biochemistry, Core Facility, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Andreas Bracher
- The Max-Planck Institute of Biochemistry, Department of Cellular Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| | - Wolfgang Baumeister
- The Max-Planck Institute of Biochemistry, Department of Molecular Structural Biology, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|
119
|
Daino GL, Frau A, Sanna C, Rigano D, Distinto S, Madau V, Esposito F, Fanunza E, Bianco G, Taglialatela-Scafati O, Zinzula L, Maccioni E, Corona A, Tramontano E. Identification of Myricetin as an Ebola Virus VP35-Double-Stranded RNA Interaction Inhibitor through a Novel Fluorescence-Based Assay. Biochemistry 2018; 57:6367-6378. [PMID: 30298725 DOI: 10.1021/acs.biochem.8b00892] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ebola virus (EBOV) is a filovirus that causes a severe and rapidly progressing hemorrhagic syndrome; a recent epidemic illustrated the urgent need for novel therapeutic agents because no drugs have been approved for treatment of Ebola virus. A key contribution to the high lethality observed during EBOV outbreaks comes from viral evasion of the host antiviral innate immune response in which viral protein VP35 plays a crucial role, blocking interferon type I production, first by masking the viral double-stranded RNA (dsRNA) and preventing its detection by the pattern recognition receptor RIG-I. Aiming to identify inhibitors of the interaction of VP35 with the viral dsRNA, counteracting the VP35 viral innate immune evasion, we established a new methodology for high-yield recombinant VP35 (rVP35) expression and purification and a novel and robust fluorescence-based rVP35-RNA interaction assay ( Z' factor of 0.69). Taking advantage of such newly established methods, we screened a small library of Sardinian natural extracts, identifying Limonium morisianum as the most potent inhibitor extract. A bioguided fractionation led to the identification of myricetin as the component that can inhibit rVP35-dsRNA interaction with an IC50 value of 2.7 μM. Molecular docking studies showed that myricetin interacts with the highly conserved region of the VP35 RNA binding domain, laying the basis for further structural optimization of potent inhibitors of VP35-dsRNA interaction.
Collapse
Affiliation(s)
- Gian Luca Daino
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Aldo Frau
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Cinzia Sanna
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Daniela Rigano
- Department of Pharmacy, School of Medicine and Surgery , University of Naples Federico II , Naples 80131 , Italy
| | - Simona Distinto
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Veronica Madau
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Francesca Esposito
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Elisa Fanunza
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Giulia Bianco
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Orazio Taglialatela-Scafati
- Department of Pharmacy, School of Medicine and Surgery , University of Naples Federico II , Naples 80131 , Italy
| | - Luca Zinzula
- The Max-Planck Institute of Biochemistry , Department of Molecular Structural Biology , Martinsried 82152 , Germany
| | - Elias Maccioni
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Angela Corona
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences , University of Cagliari , Cagliari 09042 , Italy.,Istituto di Ricerca Genetica e Biomedica , Consiglio Nazionale delle Ricerche (CNR) , Monserrato 09042 , Italy
| |
Collapse
|
120
|
Drug Repurposing for Ebola Virus Disease: Principles of Consideration and the Animal Rule. J Pharm Sci 2018; 108:798-806. [PMID: 30244014 DOI: 10.1016/j.xphs.2018.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/29/2018] [Accepted: 09/11/2018] [Indexed: 11/21/2022]
Abstract
There are no approved drugs or biologics to treat Ebola virus disease (EVD). Literature reviews identified a list of 141 drugs with reports of preliminary in vitro potency and in vivo effectiveness in animals or with reports of clinical use/trials in EVD patients. The majority of these drugs have been individually approved by the U.S. Food and Drug Administration for treating various non-EVD diseases. The anti-Ebola potency data of these drugs were curated from literature and publicly accessible databases, along with their individual biopharmaceutical and pharmacokinetic characteristics. To facilitate the development of antiviral drugs including anti-EVD drugs, highlights include optimization of the exposure-response relationship, design of a safe and effective clinical dosing regimen to achieve an adequate high ratio of clinical Cmin to a plasma protein binding-adjusted EC95, and the pharmacokinetic studies needed in animal models (healthy and affected) and in healthy volunteers. The exposure/response relationship for human dose selection is summarized, as described in the U.S. Food and Drug Administration "Animal Rule'' guidance when human efficacy studies are not ethical or feasible.
Collapse
|
121
|
Cong VT, Gaus K, Tilley RD, Gooding JJ. Rod-shaped mesoporous silica nanoparticles for nanomedicine: recent progress and perspectives. Expert Opin Drug Deliv 2018; 15:881-892. [PMID: 30173560 DOI: 10.1080/17425247.2018.1517748] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Interest in mesoporous silica nanoparticles for drug delivery has resulted in a good understanding of the impact of size and surface chemistry of these nanoparticles on their performance as drug carriers. Shape has emerged as an additional factor that can have a significant effect on delivery efficacy. Rod-shaped mesoporous silica nanoparticles show improvements in drug delivery relative to spherical mesoporous silica nanoparticles. AREAS COVERED This review summarises the synthesis methods for producing rod-shaped mesoporous silica nanoparticles for use in nanomedicine. The second part covers recent progress of mesoporous silica nanorods by comparing the impact of sphere and rod-shape on drug delivery efficiency. EXPERT OPINION As hollow mesoporous silica nanorods are capable of higher drug loads than most other drug delivery vehicles, such particles will reduce the amount of mesoporous silica in the body for efficient therapy. However, the importance of nanoparticle shape on drug delivery efficiency is not well understood for mesoporous silica. Studies that visualize and quantify the uptake pathway of mesoporous silica nanorods in specific cell types and compare the cellular uptake to the well-studied nanospheres should be the focus of research to better understand the role of shape in uptake.
Collapse
Affiliation(s)
- Vu Thanh Cong
- a School of Chemistry, Australian of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , University of New South Wales , Sydney , Australia
| | - Katharina Gaus
- b EMBL Australia Node in Single Molecule Science and ARC Centre of Excellence in Advanced Molecular Imaging , University of New South Wales , Sydney , Australia
| | - Richard D Tilley
- a School of Chemistry, Australian of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , University of New South Wales , Sydney , Australia
| | - J Justin Gooding
- a School of Chemistry, Australian of NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , University of New South Wales , Sydney , Australia
| |
Collapse
|
122
|
Luthra P, Naidoo J, Pietzsch CA, De S, Khadka S, Anantpadma M, Williams CG, Edwards MR, Davey RA, Bukreyev A, Ready JM, Basler CF. Inhibiting pyrimidine biosynthesis impairs Ebola virus replication through depletion of nucleoside pools and activation of innate immune responses. Antiviral Res 2018; 158:288-302. [PMID: 30144461 DOI: 10.1016/j.antiviral.2018.08.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/16/2018] [Accepted: 08/18/2018] [Indexed: 12/27/2022]
Abstract
Specific host pathways that may be targeted therapeutically to inhibit the replication of Ebola virus (EBOV) and other emerging viruses remain incompletely defined. A screen of 200,000 compounds for inhibition of an EBOV minigenome (MG) assay that measures the function of the viral polymerase complex identified as hits several compounds with an amino-tetrahydrocarbazole scaffold. This scaffold was structurally similar to GSK983, a compound previously described as having broad-spectrum antiviral activity due to its impairing de novo pyrimidine biosynthesis through inhibition of dihydroorotate dehydrogenase (DHODH). We generated compound SW835, the racemic version of GSK983 and demonstrated that SW835 and brequinar, another DHODH inhibitor, potently inhibit the MG assay and the replication of EBOV, vesicular stomatitis virus (VSV) and Zika (ZIKV) in vitro. Nucleoside and deoxynucleoside supplementation studies demonstrated that depletion of pyrimidine pools contributes to antiviral activity of these compounds. As reported for other DHODH inhibitors, SW835 and brequinar also induced expression of interferon stimulated genes (ISGs). ISG induction was demonstrated to occur without production of IFNα/β and independently of the IFNα receptor and was not blocked by EBOV-encoded suppressors of IFN signaling pathways. Furthermore, we demonstrated that transcription factor IRF1 is required for this ISG induction, and that IRF1 induction requires the DNA damage response kinase ATM. Therefore, de novo pyrimidine biosynthesis is critical for the replication of EBOV and other RNA viruses and inhibition of this pathway activates an ATM and IRF1-dependent innate immune response that subverts EBOV immune evasion functions.
Collapse
Affiliation(s)
- Priya Luthra
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Jacinth Naidoo
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Colette A Pietzsch
- Department of Pathology, Galveston National Laboratory, The University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Sampriti De
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sudip Khadka
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Manu Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Caroline G Williams
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Megan R Edwards
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Robert A Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Alexander Bukreyev
- Department of Pathology, Galveston National Laboratory, The University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA; Department of Microbiology & Immunology, Galveston National Laboratory, The University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Joseph M Ready
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
123
|
Kuzmina NA, Younan P, Gilchuk P, Santos RI, Flyak AI, Ilinykh PA, Huang K, Lubaki NM, Ramanathan P, Crowe JE, Bukreyev A. Antibody-Dependent Enhancement of Ebola Virus Infection by Human Antibodies Isolated from Survivors. Cell Rep 2018; 24:1802-1815.e5. [PMID: 30110637 PMCID: PMC6697154 DOI: 10.1016/j.celrep.2018.07.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 06/12/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022] Open
Abstract
Some monoclonal antibodies (mAbs) recovered from survivors of filovirus infections can protect against infection. It is currently unknown whether natural infection also induces some antibodies with the capacity for antibody-dependent enhancement (ADE). A panel of mAbs obtained from human survivors of filovirus infection caused by Ebola, Bundibugyo, or Marburg viruses was evaluated for their ability to facilitate ADE. ADE was observed readily with all mAbs examined at sub-neutralizing concentrations, and this effect was not restricted to mAbs with a particular epitope specificity, neutralizing capacity, or subclass. Blocking of specific Fcγ receptors reduced but did not abolish ADE that was associated with high-affinity binding antibodies, suggesting that lower-affinity interactions still cause ADE. Mutations of Fc fragments of an mAb that altered its interaction with Fc receptors rendered the antibody partially protective in vivo at a low dose, suggesting that ADE counteracts antibody-mediated protection and facilitates dissemination of filovirus infections.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/pharmacology
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Neutralizing/pharmacology
- Antibodies, Viral/isolation & purification
- Antibodies, Viral/pharmacology
- Antibody-Dependent Enhancement
- Ebolavirus/drug effects
- Ebolavirus/genetics
- Ebolavirus/immunology
- Ebolavirus/pathogenicity
- Epitopes/genetics
- Epitopes/immunology
- Gene Expression
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/mortality
- Hemorrhagic Fever, Ebola/therapy
- Hemorrhagic Fever, Ebola/virology
- Humans
- Immune Sera/chemistry
- Immunoglobulin Fc Fragments/chemistry
- Immunoglobulin Fc Fragments/genetics
- Marburg Virus Disease/immunology
- Marburg Virus Disease/mortality
- Marburg Virus Disease/therapy
- Marburg Virus Disease/virology
- Marburgvirus/drug effects
- Marburgvirus/genetics
- Marburgvirus/pathogenicity
- Mice
- Mice, Inbred BALB C
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/virology
- Primary Cell Culture
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Survival Analysis
- Survivors
- THP-1 Cells
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Patrick Younan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rodrigo I Santos
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Andrew I Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Ndongala M Lubaki
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Palaniappan Ramanathan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
124
|
Rogers KJ, Maury W. The role of mononuclear phagocytes in Ebola virus infection. J Leukoc Biol 2018; 104:717-727. [PMID: 30095866 DOI: 10.1002/jlb.4ri0518-183r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 12/17/2022] Open
Abstract
The filovirus, Zaire Ebolavirus (EBOV), infects tissue macrophages (Mϕs) and dendritic cells (DCs) early during infection. Viral infection of both cells types is highly productive, leading to increased viral load. However, virus infection of these two cell types results in different consequences for cellular function. Infection of Mϕs stimulates the production of proinflammatory and immunomodulatory cytokines and chemokines, leading to the production of a cytokine storm, while simultaneously increasing tissue factor production and thus facilitating disseminated intravascular coagulation. In contrast, EBOV infection of DCs blocks DC maturation and antigen presentation rendering these cells unable to communicate with adaptive immune response elements. Details of the known interactions of these cells with EBOV are reviewed here. We also identify a number of unanswered questions that remain about interactions of filoviruses with these cells.
Collapse
Affiliation(s)
- Kai J Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
125
|
Martin B, Decroly É. Mécanismes d’échappement des filovirus à l’immunité innée. Med Sci (Paris) 2018; 34:671-677. [DOI: 10.1051/medsci/20183408013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Le virus Ébola est un pathogène émergent important en Afrique où il a été responsable de plusieurs épidémies de fièvres hémorragiques associées à un taux de mortalité extrêmement élevée (jusqu’à 90 %). La pathogenèse des filovirus est, entre autres, liée à une réponse antivirale inadaptée. Cette famille de virus a en effet développé des stratégies d’échappement aux mécanismes précoces de l’immunité innée. Il en résulte une réplication virale massive qui induit une réponse immunitaire inappropriée à l’origine d’une réaction inflammatoire aiguë associée au syndrome hémorragique. Dans cette revue, nous décrivons les mécanismes utilisés par les filovirus, tels que le virus Ébola, pour échapper à la réponse immunitaire innée.
Collapse
|
126
|
Leiva-Suero LE, Morales JM, Villacís-Valencia SE, Escalona-Rabaza M, Quishpe-Jara GDLM, Hernández-Navarro EV, Fernández-Nieto M. Ébola, abordaje clínico integral. REVISTA DE LA FACULTAD DE MEDICINA 2018. [DOI: 10.15446/revfacmed.v66n3.64545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introducción. El virus del Ébola, antes llamado fiebre hemorrágica del Ébola, es una enfermedad altamente contagiosa con mortalidad entre 50% y 90%, para la cual existen prometedoras opciones de tratamiento que se encuentran en fase de evaluación y uso compasional.Objetivos. Revisar la mejor evidencia médica publicada y analizar el comportamiento de las epidemias por virus del Ébola, sus manifestaciones clínicas, sus complicaciones, los elementos más significativos para su diagnóstico y las nuevas opciones terapéuticas disponibles, para así aprender y aplicar estas experiencias en nuevos brotes.Materiales y métodos. Se realizó una búsqueda sistemática en las bases de datos PubMed, ProQuest, Embase, Redalyc, Ovid, Medline, DynaMed y ClinicalKey durante el periodo 2009-2017 en el contexto internacional, regional y local.Resultados. La revisión sistemática de artículos aportó un total de 51 430 registros, de los cuales 772 eran elegibles; de estos, 722 no eran relevantes, por lo que quedaron incluidos 50. A punto de partida se pudieron precisar los aspectos objeto de esta revisión.Conclusión. La enfermedad causada por el virus del Ébola, a pesar de su alta mortalidad, puede ser prevenida, diagnosticada oportunamente y tratada con efectividad, lo cual permite evaluar su impacto epidemiológico en las áreas endémicas y a nivel mundial. Existe un potencial arsenal terapéutico en fase de experimentación con resultados prometedores.
Collapse
|
127
|
Filovirus – Auslöser von hämorrhagischem Fieber. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2018; 61:894-907. [DOI: 10.1007/s00103-018-2757-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
128
|
Singh K, Marasini B, Chen X, Spearman P. A novel Ebola virus antibody-dependent cell-mediated cytotoxicity (Ebola ADCC) assay. J Immunol Methods 2018; 460:10-16. [PMID: 29894746 DOI: 10.1016/j.jim.2018.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/03/2018] [Accepted: 06/06/2018] [Indexed: 12/21/2022]
Abstract
Ebolaviruses are highly virulent pathogens that cause Ebola viral disease (EVD). Data from non-human primate (NHP) models and from human survivors of EVD suggest that anti-Ebola antibodies play an integral role in protection. Antibody-dependent cell-mediated cytotoxicity (ADCC) is a potential mechanism through which anti-Ebola antibodies may mediate protection. We developed a robust Ebola-specific ADCC assay for use in ongoing trials of Ebola vaccines. Stable cell lines for inducible Zaire ebolavirus glycoprotein (EBOV GP) expression were developed to provide a uniform source of target cells in the assay, and were combined with an existing human natural killer (NK) cell line as the effector cell. When applied to commercially available anti-EBOV GP monoclonal antibodies, the assay clearly differentiated antibody with high ADCC activity from those with low or no ADCC activity. Anti-EBOV ADCC activity was also detected in plasma samples from rhesus macaques immunized with a candidate Ebola vaccine. The Ebola ADCC assay reported here will be a useful tool in studying the functionality of anti-EBOV GP antibodies elicited by Ebola vaccines in ongoing and future clinical trials.
Collapse
Affiliation(s)
- Karnail Singh
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Bishal Marasini
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Xuemin Chen
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Paul Spearman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
129
|
A GXXXA Motif in the Transmembrane Domain of the Ebola Virus Glycoprotein Is Required for Tetherin Antagonism. J Virol 2018; 92:JVI.00403-18. [PMID: 29669839 DOI: 10.1128/jvi.00403-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/13/2018] [Indexed: 02/04/2023] Open
Abstract
The interferon-induced antiviral host cell protein tetherin can inhibit the release of several enveloped viruses from infected cells. The Ebola virus (EBOV) glycoprotein (GP) antagonizes tetherin, but the domains and amino acids in GP that are required for tetherin antagonism have not been fully defined. A GXXXA motif within the transmembrane domain (TMD) of EBOV-GP was previously shown to be important for GP-mediated cellular detachment. Here, we investigated whether this motif also contributes to tetherin antagonism. Mutation of the GXXXA motif did not impact GP expression or particle incorporation and only modestly reduced EBOV-GP-driven entry. In contrast, the GXXXA motif was required for tetherin antagonism in transfected cells. Moreover, alteration of the GXXXA motif increased tetherin sensitivity of a replication-competent vesicular stomatitis virus (VSV) chimera encoding EBOV-GP. Although these results await confirmation with authentic EBOV, they indicate that a GXXXA motif in the TMD of EBOV-GP is important for tetherin antagonism. Moreover, they provide the first evidence that GP can antagonize tetherin in the context of an infectious EBOV surrogate.IMPORTANCE The glycoprotein (GP) of Ebola virus (EBOV) inhibits the antiviral host cell protein tetherin and may promote viral spread in tetherin-positive cells. However, tetherin antagonism by GP has so far been demonstrated only with virus-like particles, and it is unknown whether GP can block tetherin in infected cells. Moreover, a mutation in GP that selectively abrogates tetherin antagonism is unknown. Here, we show that a GXXXA motif in the transmembrane domain of EBOV-GP, which was previously reported to be required for GP-mediated cell rounding, is also important for tetherin counteraction. Moreover, analysis of this mutation in the context of vesicular stomatitis virus chimeras encoding EBOV-GP revealed that GP-mediated tetherin counteraction is operative in infected cells. To our knowledge, these findings demonstrate for the first time that GP can antagonize tetherin in infected cells and provide a tool to study the impact of GP-dependent tetherin counteraction on EBOV spread.
Collapse
|
130
|
Desrochers GF, Cornacchia C, McKay CS, Pezacki JP. Activity-Based Phosphatidylinositol Kinase Probes Detect Changes to Protein-Protein Interactions During Hepatitis C Virus Replication. ACS Infect Dis 2018; 4:752-757. [PMID: 29509402 DOI: 10.1021/acsinfecdis.8b00047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Protein-protein interactions are integral to host-virus interactions and can contribute significantly to enzyme regulation by changing the localization of both host and viral enzymes within the cell, inducing conformational change relevant to enzyme activity or recruiting other additional proteins to form functional complexes. Identifying the interactors of active enzymes using an activity-based protein profiling probe has allowed us to characterize both normal enzyme activation mechanisms and the manner by which these mechanisms are hijacked and altered by the hepatitis C virus (HCV). Here, we report use of a novel activity-based probe, PIKBPyne, which labels phosphatidylinositol kinases (PIKs) in an activity-based manner, to investigate HCV-dependent changes in protein-protein interactions for PI4KB. Herein, we report the synthesis of new variations on PIKBPyne, compare their ability to label the interacting partners of PI4KB, and demonstrate the utility of our approach in characterizing virus-mediated changes to host function.
Collapse
Affiliation(s)
- Geneviève F. Desrochers
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Pvt., Ottawa, Ontario K1N 6N5, Canada
| | - Christina Cornacchia
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Pvt., Ottawa, Ontario K1N 6N5, Canada
| | - Craig S. McKay
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Pvt., Ottawa, Ontario K1N 6N5, Canada
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Pvt., Ottawa, Ontario K1N 6N5, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Rd., Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
131
|
Affiliation(s)
- Patrick Younan
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Mathieu Iampietro
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Department Microbiology & Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| |
Collapse
|
132
|
Su Z, Wu C, Shi L, Luthra P, Pintilie GD, Johnson B, Porter JR, Ge P, Chen M, Liu G, Frederick TE, Binning JM, Bowman GR, Zhou ZH, Basler CF, Gross ML, Leung DW, Chiu W, Amarasinghe GK. Electron Cryo-microscopy Structure of Ebola Virus Nucleoprotein Reveals a Mechanism for Nucleocapsid-like Assembly. Cell 2018; 172:966-978.e12. [PMID: 29474922 PMCID: PMC5973842 DOI: 10.1016/j.cell.2018.02.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 12/27/2017] [Accepted: 01/31/2018] [Indexed: 01/24/2023]
Abstract
Ebola virus nucleoprotein (eNP) assembles into higher-ordered structures that form the viral nucleocapsid (NC) and serve as the scaffold for viral RNA synthesis. However, molecular insights into the NC assembly process are lacking. Using a hybrid approach, we characterized the NC-like assembly of eNP, identified novel regulatory elements, and described how these elements impact function. We generated a three-dimensional structure of the eNP NC-like assembly at 5.8 Å using electron cryo-microscopy and identified a new regulatory role for eNP helices α22-α23. Biochemical, biophysical, and mutational analyses revealed that inter-eNP contacts within α22-α23 are critical for viral NC assembly and regulate viral RNA synthesis. These observations suggest that the N terminus and α22-α23 of eNP function as context-dependent regulatory modules (CDRMs). Our current study provides a framework for a structural mechanism for NC-like assembly and a new therapeutic target.
Collapse
Affiliation(s)
- Zhaoming Su
- Department of Bioengineering and Department of Microbiology and Immunology, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Chao Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Liuqing Shi
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Priya Luthra
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Grigore D Pintilie
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Britney Johnson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Justin R Porter
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Peng Ge
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Muyuan Chen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gai Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas E Frederick
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer M Binning
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Z Hong Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wah Chiu
- Department of Bioengineering and Department of Microbiology and Immunology, James H. Clark Center, Stanford University, Stanford, CA 94305, USA.
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
133
|
Insights into the homo-oligomerization properties of N-terminal coiled-coil domain of Ebola virus VP35 protein. Virus Res 2018; 247:61-70. [PMID: 29427597 DOI: 10.1016/j.virusres.2018.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/30/2018] [Accepted: 02/06/2018] [Indexed: 12/31/2022]
Abstract
The multifunctional Ebola virus (EBOV) VP35 protein is a key determinant of virulence. VP35 is essential for EBOV replication, is a component of the viral RNA polymerase and participates in nucleocapsid formation. Furthermore, VP35 contributes to EBOV evasion of the host innate immune response by suppressing RNA silencing and blocking RIG-I like receptors' pathways that lead to type I interferon (IFN) production. VP35 homo-oligomerization has been reported to be critical for its replicative function and to increase its IFN-antagonism properties. Moreover, homo-oligomerization is mediated by a predicted coiled-coil (CC) domain located within its N-terminal region. Here we report the homo-oligomerization profile of full-length recombinant EBOV VP35 (rVP35) assessed by size-exclusion chromatography and native polyacrylamide gel electrophoresis. Based on our biochemical results and in agreement with previous experimental observations, we have built an in silico 3D model of the so-far structurally unsolved EBOV VP35 CC domain and performed self-assembly homo-oligomerization simulations by means of molecular dynamics. Our model advances the understanding of how VP35 may associate in different homo-oligomeric species, a crucial process for EBOV replication and pathogenicity.
Collapse
|
134
|
Luthra P, Liang J, Pietzsch CA, Khadka S, Edwards MR, Wei S, De S, Posner B, Bukreyev A, Ready JM, Basler CF. A high throughput screen identifies benzoquinoline compounds as inhibitors of Ebola virus replication. Antiviral Res 2018; 150:193-201. [PMID: 29294299 PMCID: PMC11727378 DOI: 10.1016/j.antiviral.2017.12.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/22/2017] [Accepted: 12/26/2017] [Indexed: 01/28/2023]
Abstract
Ebola virus (EBOV) is an enveloped negative-sense, single-stranded RNA virus of the filovirus family that causes severe disease in humans. Approved therapies for EBOV disease are lacking. EBOV RNA synthesis is carried out by a virus-encoded complex with RNA-dependent RNA polymerase activity that is required for viral propagation. This complex and its activities are therefore potential antiviral targets. To identify potential lead inhibitors of EBOV RNA synthesis, a library of small molecule compounds was screened against a previously established assay of EBOV RNA synthesis, the EBOV minigenome assay (MGA), in 384 well microplate format. The screen identified 56 hits that inhibited EBOV MGA activity by more than 70% while exhibiting less than 20% cell cytotoxicity. Inhibitory chemical scaffolds included angelicin derivatives, derivatives of the antiviral compound GSK983 and benzoquinolines. Structure-activity relationship (SAR) studies of the benzoquinoline scaffold produced ∼50 analogs and led to identification of an optimized compound, SW456, with a submicromolar IC50 in the EBOV MGA and antiviral activity against infectious EBOV in cell culture. The compound was also active against a MGA for another deadly filovirus, Marburg virus. It also exhibited antiviral activity towards a negative-sense RNA virus from the rhabdovirus family, vesicular stomatitis virus, and a positive-sense RNA virus, Zika virus. Overall, these data demonstrate the potential of the EBOV MGA to identify anti-EBOV compounds and identifies the benzoquinoline series as a broad-spectrum antiviral lead.
Collapse
Affiliation(s)
- Priya Luthra
- Center of Microbial Pathogenesis, Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Jue Liang
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA
| | - Colette A Pietzsch
- Department of Pathology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Sudip Khadka
- Center of Microbial Pathogenesis, Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Megan R Edwards
- Center of Microbial Pathogenesis, Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Shuguang Wei
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA
| | - Sampriti De
- Center of Microbial Pathogenesis, Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA; Department of Microbiology & Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA
| | - Christopher F Basler
- Center of Microbial Pathogenesis, Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
135
|
McElroy AK, Mühlberger E, Muñoz-Fontela C. Immune barriers of Ebola virus infection. Curr Opin Virol 2018; 28:152-160. [PMID: 29452995 PMCID: PMC5886007 DOI: 10.1016/j.coviro.2018.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 01/10/2023]
Abstract
Since its initial emergence in 1976 in northern Democratic Republic of Congo (DRC), Ebola virus (EBOV) has been a global health concern due to its virulence in humans, the mystery surrounding the identity of its host reservoir and the unpredictable nature of Ebola virus disease (EVD) outbreaks. Early after the first clinical descriptions of a disease resembling a 'septic-shock-like syndrome', with coagulation abnormalities and multi-system organ failure, researchers began to evaluate the role of the host immune response in EVD pathophysiology. In this review, we summarize how data gathered during the last 40 years in the laboratory as well as in the field have provided insight into EBOV immunity. From molecular mechanisms involved in EBOV recognition in infected cells, to antigen processing and adaptive immune responses, we discuss current knowledge on the main immune barriers of infection as well as outstanding research questions.
Collapse
Affiliation(s)
- Anita K McElroy
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, 3501 Fifth Ave, Pittsburgh, PA 15261, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, 02118 Boston, MA, USA
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Strasse 74, 20359 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg, Germany.
| |
Collapse
|
136
|
|
137
|
Ren S, Wei Q, Cai L, Yang X, Xing C, Tan F, Leavenworth JW, Liang S, Liu W. Alphavirus Replicon DNA Vectors Expressing Ebola GP and VP40 Antigens Induce Humoral and Cellular Immune Responses in Mice. Front Microbiol 2018; 8:2662. [PMID: 29375526 PMCID: PMC5767729 DOI: 10.3389/fmicb.2017.02662] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/20/2017] [Indexed: 11/13/2022] Open
Abstract
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and no approved therapeutics or vaccine is currently available. Glycoprotein (GP) is the major protective antigen of EBOV, and can generate virus-like particles (VLPs) by co-expression with matrix protein (VP40). In this study, we constructed a recombinant Alphavirus Semliki Forest virus (SFV) replicon vector DREP to express EBOV GP and matrix viral protein (VP40). EBOV VLPs were successfully generated and achieved budding from 293 cells after co-transfection with DREP-based GP and VP40 vectors (DREP-GP+DREP-VP40). Vaccination of BALB/c mice with DREP-GP, DREP-VP40, or DREP-GP+DREP-VP40 vectors, followed by immediate electroporation resulted in a mixed IgG subclass production, which recognized EBOV GP and/or VP40 proteins. This vaccination regimen also led to the generation of both Th1 and Th2 cellular immune responses in mice. Notably, vaccination with DREP-GP and DREP-VP40, which produces both GP and VP40 antigens, induced a significantly higher level of anti-GP IgG2a antibody and increased IFN-γ secreting CD8+ T-cell responses relative to vaccination with DREP-GP or DREP-VP40 vector alone. Our study indicates that co-expression of GP and VP40 antigens based on the SFV replicon vector generates EBOV VLPs in vitro, and vaccination with recombinant DREP vectors containing GP and VP40 antigens induces Ebola antigen-specific humoral and cellular immune responses in mice. This novel approach provides a simple and efficient vaccine platform for Ebola disease prevention.
Collapse
Affiliation(s)
- Shoufeng Ren
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China
| | - Qimei Wei
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Liya Cai
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Xuejing Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuicui Xing
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China
| | - Feng Tan
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shaohui Liang
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Wenquan Liu
- Department of Human Parasitology, Wenzhou Medical University, Wenzhou, China.,Institute of Pathogen and Immunology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
138
|
Dutta M, Robertson SJ, Okumura A, Scott DP, Chang J, Weiss JM, Sturdevant GL, Feldmann F, Haddock E, Chiramel AI, Ponia SS, Dougherty JD, Katze MG, Rasmussen AL, Best SM. A Systems Approach Reveals MAVS Signaling in Myeloid Cells as Critical for Resistance to Ebola Virus in Murine Models of Infection. Cell Rep 2017; 18:816-829. [PMID: 28099857 DOI: 10.1016/j.celrep.2016.12.069] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/11/2016] [Accepted: 12/20/2016] [Indexed: 01/08/2023] Open
Abstract
The unprecedented 2013-2016 outbreak of Ebola virus (EBOV) resulted in over 11,300 human deaths. Host resistance to RNA viruses requires RIG-I-like receptor (RLR) signaling through the adaptor protein, mitochondrial antiviral signaling protein (MAVS), but the role of RLR-MAVS in orchestrating anti-EBOV responses in vivo is not known. Here we apply a systems approach to MAVS-/- mice infected with either wild-type or mouse-adapted EBOV. MAVS controlled EBOV replication through the expression of IFNα, regulation of inflammatory responses in the spleen, and prevention of cell death in the liver, with macrophages implicated as a major cell type influencing host resistance. A dominant role for RLR signaling in macrophages was confirmed following conditional MAVS deletion in LysM+ myeloid cells. These findings reveal tissue-specific MAVS-dependent transcriptional pathways associated with resistance to EBOV, and they demonstrate that EBOV adaptation to cause disease in mice involves changes in two distinct events, RLR-MAVS antagonism and suppression of RLR-independent IFN-I responses.
Collapse
Affiliation(s)
- Mukta Dutta
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 59105, USA
| | - Shelly J Robertson
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Atsushi Okumura
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 59105, USA; Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA; Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Dana P Scott
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Jean Chang
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 59105, USA
| | - Jeffrey M Weiss
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 59105, USA
| | - Gail L Sturdevant
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Friederike Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Elaine Haddock
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Abhilash I Chiramel
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Sanket S Ponia
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Jonathan D Dougherty
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Michael G Katze
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 59105, USA
| | - Angela L Rasmussen
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 59105, USA; Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Sonja M Best
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA.
| |
Collapse
|
139
|
Eisfeld AJ, Halfmann PJ, Wendler JP, Kyle JE, Burnum-Johnson KE, Peralta Z, Maemura T, Walters KB, Watanabe T, Fukuyama S, Yamashita M, Jacobs JM, Kim YM, Casey CP, Stratton KG, Webb-Robertson BJM, Gritsenko MA, Monroe ME, Weitz KK, Shukla AK, Tian M, Neumann G, Reed JL, van Bakel H, Metz TO, Smith RD, Waters KM, N'jai A, Sahr F, Kawaoka Y. Multi-platform 'Omics Analysis of Human Ebola Virus Disease Pathogenesis. Cell Host Microbe 2017; 22:817-829.e8. [PMID: 29154144 DOI: 10.1016/j.chom.2017.10.011] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/13/2017] [Accepted: 09/20/2017] [Indexed: 12/11/2022]
Abstract
The pathogenesis of human Ebola virus disease (EVD) is complex. EVD is characterized by high levels of virus replication and dissemination, dysregulated immune responses, extensive virus- and host-mediated tissue damage, and disordered coagulation. To clarify how host responses contribute to EVD pathophysiology, we performed multi-platform 'omics analysis of peripheral blood mononuclear cells and plasma from EVD patients. Our results indicate that EVD molecular signatures overlap with those of sepsis, imply that pancreatic enzymes contribute to tissue damage in fatal EVD, and suggest that Ebola virus infection may induce aberrant neutrophils whose activity could explain hallmarks of fatal EVD. Moreover, integrated biomarker prediction identified putative biomarkers from different data platforms that differentiated survivors and fatalities early after infection. This work reveals insight into EVD pathogenesis, suggests an effective approach for biomarker identification, and provides an important community resource for further analysis of human EVD severity.
Collapse
Affiliation(s)
- Amie J Eisfeld
- Department of Pathobiological Sciences, University of Wisconsin - Madison (UW-Madison), Madison, WI 53706, USA
| | - Peter J Halfmann
- Department of Pathobiological Sciences, University of Wisconsin - Madison (UW-Madison), Madison, WI 53706, USA
| | - Jason P Wendler
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Kristin E Burnum-Johnson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Zuleyma Peralta
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai (ISMMS), New York City, NY 10029, USA
| | - Tadashi Maemura
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science (IMS), University of Tokyo, Tokyo 108-8639, Japan
| | - Kevin B Walters
- Department of Pathobiological Sciences, University of Wisconsin - Madison (UW-Madison), Madison, WI 53706, USA
| | - Tokiko Watanabe
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science (IMS), University of Tokyo, Tokyo 108-8639, Japan
| | - Satoshi Fukuyama
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science (IMS), University of Tokyo, Tokyo 108-8639, Japan
| | - Makoto Yamashita
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science (IMS), University of Tokyo, Tokyo 108-8639, Japan
| | - Jon M Jacobs
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Young-Mo Kim
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Cameron P Casey
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Kelly G Stratton
- Computing and Analytics Division, National Security Directorate, PNNL, Richland, WA 99352, USA
| | | | - Marina A Gritsenko
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Matthew E Monroe
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Karl K Weitz
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Anil K Shukla
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA
| | - Mingyuan Tian
- Department of Chemical and Biological Engineering, UW-Madison, Madison, WI 53706, USA
| | - Gabriele Neumann
- Department of Pathobiological Sciences, University of Wisconsin - Madison (UW-Madison), Madison, WI 53706, USA
| | - Jennifer L Reed
- Department of Chemical and Biological Engineering, UW-Madison, Madison, WI 53706, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai (ISMMS), New York City, NY 10029, USA; Icahn Institute for Genomics and Multiscale Biology, ISMMS, New York City, NY 10029, USA.
| | - Thomas O Metz
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA.
| | - Richard D Smith
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA.
| | - Katrina M Waters
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA 99352, USA.
| | - Alhaji N'jai
- Department of Pathobiological Sciences, University of Wisconsin - Madison (UW-Madison), Madison, WI 53706, USA; Department of Biological Sciences, Fourah Bay College, College of Medicine & Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | - Foday Sahr
- 34(th) Regimental Military Hospital at Wilberforce, Freetown, Sierra Leone.
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin - Madison (UW-Madison), Madison, WI 53706, USA; Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science (IMS), University of Tokyo, Tokyo 108-8639, Japan; International Research Center for Infectious Diseases, IMS, University of Tokyo, Tokyo 108-8639, Japan.
| |
Collapse
|
140
|
Banerjee A, Pal A, Pal D, Mitra P. Ebolavirus interferon antagonists—protein interaction perspectives to combat pathogenesis. Brief Funct Genomics 2017; 17:392-401. [DOI: 10.1093/bfgp/elx034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
141
|
Speranza E, Altamura LA, Kulcsar K, Bixler SL, Rossi CA, Schoepp RJ, Nagle E, Aguilar W, Douglas CE, Delp KL, Minogue TD, Palacios G, Goff AJ, Connor JH. Comparison of Transcriptomic Platforms for Analysis of Whole Blood from Ebola-Infected Cynomolgus Macaques. Sci Rep 2017; 7:14756. [PMID: 29116224 PMCID: PMC5676990 DOI: 10.1038/s41598-017-15145-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/17/2017] [Indexed: 11/09/2022] Open
Abstract
Ebola virus disease (EVD) is a serious illness with mortality rates of 20-90% in various outbreaks. EVD is characterized by robust virus replication and strong host inflammatory response. Analyzing host immune responses has increasingly involved multimodal approaches including transcriptomics to profile gene expression. We studied cynomolgus macaques exposed to Ebola virus Makona via different routes with the intent of comparing RNA-Seq to a NanoString nCounter codeset targeting 769 non-human primate (NHP) genes. RNA-Seq analysis of serial blood samples showed different routes led to the same overall transcriptional response seen in previously reported EBOV-exposed NHP studies. Both platforms displayed a strong correlation in gene expression patterns, including a strong induction of innate immune response genes at early times post-exposure, and neutrophil-associated genes at later time points. A 41-gene classifier was tested in both platforms for ability to cluster samples by infection status. Both NanoString and RNA-Seq could be used to predict relative abundances of circulating immune cell populations that matched traditional hematology. This demonstrates the complementarity of RNA-Seq and NanoString. Moreover, the development of an NHP-specific NanoString codeset should augment studies of filoviruses and other high containment infectious diseases without the infrastructure requirements of RNA-Seq technology.
Collapse
Affiliation(s)
- Emily Speranza
- Department of Microbiology, Bioinformatics Program, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, United States
| | - Louis A Altamura
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Kirsten Kulcsar
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Sandra L Bixler
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Cynthia A Rossi
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Randal J Schoepp
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Elyse Nagle
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - William Aguilar
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Christina E Douglas
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Korey L Delp
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Timothy D Minogue
- Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Gustavo Palacios
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Arthur J Goff
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States.
| | - John H Connor
- Department of Microbiology, Bioinformatics Program, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, United States.
| |
Collapse
|
142
|
Weiwei G, Xuexing Z, Chong W, Yongkun Z, Qi W, Hualei W, Gary W, Ying X, Haijun W, Zengguo C, Na F, Hang C, Tiecheng W, Yuwei G, Junjie S, Songtao Y, Xianzhu X. Marburg virus-like particles produced in insect cells induce neutralizing antibodies in rhesus macaques. J Med Virol 2017; 89:2069-2074. [DOI: 10.1002/jmv.24832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/06/2017] [Accepted: 04/06/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Gai Weiwei
- College of Veterinary Medicine; Jilin University; Changchun China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Zheng Xuexing
- School of Public Health; Shandong University; Jinan China
| | - Wang Chong
- State Key Laboratory of Veterinary Biotechnology; Harbin Veterinary Research Institute; Chinese Academy of Agricultural Sciences; Harbin China
| | - Zhao Yongkun
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Wang Qi
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
- College of Veterinary Medicine; Jilin Agriculture University; Changchun China
| | - Wang Hualei
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Wong Gary
- CAS Key Laboratory of Pathogenic Microbiology and Immunology; Institute of Microbiology, Chinese Academy of Sciences; Beijing China
| | - Xie Ying
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Wang Haijun
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Cao Zengguo
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Feng Na
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Chi Hang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Wang Tiecheng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Gao Yuwei
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Shan Junjie
- Institute of Pharmacology and Toxicology; Academy of Military Medical Sciences; Beijing China
| | - Yang Songtao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| | - Xia Xianzhu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control; Institute of Military Veterinary; Academy of Military Medical Sciences; Changchun China
| |
Collapse
|
143
|
He F, Melén K, Maljanen S, Lundberg R, Jiang M, Österlund P, Kakkola L, Julkunen I. Ebolavirus protein VP24 interferes with innate immune responses by inhibiting interferon-λ1 gene expression. Virology 2017; 509:23-34. [PMID: 28595092 DOI: 10.1016/j.virol.2017.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/25/2017] [Accepted: 06/01/2017] [Indexed: 10/19/2022]
Abstract
Ebolaviruses (EBOV) cause severe disease with a recent outbreak in West Africa in 2014-2015 leading to more than 28 000 cases and 11 300 fatalities. This emphasizes the urgent need for better knowledge on these highly pathogenic RNA viruses. Host innate immune responses play a key role in restricting the spread of a viral disease. In this study we systematically analyzed the effects of cloned EBOV genes on the main host immune response to RNA viruses: the activation of RIG-I pathway and type I and III interferon (IFN) gene expression. EBOV VP24, in addition of inhibiting IFN-induced antiviral responses, was found to efficiently inhibit type III IFN-λ1 gene expression. This inhibition was found to occur downstream of IRF3 activation and to be dependent on VP24 importin binding residues. These results emphasize the importance of VP24 in EBOV infection cycle, making VP24 as an excellent target for drug development.
Collapse
Affiliation(s)
- Felix He
- Institute of Biomedicine/Virology, University of Turku, Kiinamyllynkatu 13, 20520 Turku, Finland.
| | - Krister Melén
- Institute of Biomedicine/Virology, University of Turku, Kiinamyllynkatu 13, 20520 Turku, Finland; Expert Microbiology Unit, National Institute for Health and Welfare, Mannerheimintie 166, 00300 Helsinki, Finland.
| | - Sari Maljanen
- Institute of Biomedicine/Virology, University of Turku, Kiinamyllynkatu 13, 20520 Turku, Finland.
| | - Rickard Lundberg
- Institute of Biomedicine/Virology, University of Turku, Kiinamyllynkatu 13, 20520 Turku, Finland.
| | - Miao Jiang
- Expert Microbiology Unit, National Institute for Health and Welfare, Mannerheimintie 166, 00300 Helsinki, Finland.
| | - Pamela Österlund
- Expert Microbiology Unit, National Institute for Health and Welfare, Mannerheimintie 166, 00300 Helsinki, Finland.
| | - Laura Kakkola
- Institute of Biomedicine/Virology, University of Turku, Kiinamyllynkatu 13, 20520 Turku, Finland.
| | - Ilkka Julkunen
- Institute of Biomedicine/Virology, University of Turku, Kiinamyllynkatu 13, 20520 Turku, Finland.
| |
Collapse
|
144
|
The Host E3-Ubiquitin Ligase TRIM6 Ubiquitinates the Ebola Virus VP35 Protein and Promotes Virus Replication. J Virol 2017; 91:JVI.00833-17. [PMID: 28679761 DOI: 10.1128/jvi.00833-17] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/27/2017] [Indexed: 01/25/2023] Open
Abstract
Ebola virus (EBOV), a member of the Filoviridae family, is a highly pathogenic virus that causes severe hemorrhagic fever in humans and is responsible for epidemics throughout sub-Saharan, central, and West Africa. The EBOV genome encodes VP35, an important viral protein involved in virus replication by acting as an essential cofactor of the viral polymerase as well as a potent antagonist of the host antiviral type I interferon (IFN-I) system. By using mass spectrometry analysis and coimmunoprecipitation assays, we show here that VP35 is ubiquitinated on lysine 309 (K309), a residue located on its IFN antagonist domain. We also found that VP35 interacts with TRIM6, a member of the E3-ubiquitin ligase tripartite motif (TRIM) family. We recently reported that TRIM6 promotes the synthesis of unanchored K48-linked polyubiquitin chains, which are not covalently attached to any protein, to induce efficient antiviral IFN-I-mediated responses. Consistent with this notion, VP35 also associated noncovalently with polyubiquitin chains and inhibited TRIM6-mediated IFN-I induction. Intriguingly, we also found that TRIM6 enhances EBOV polymerase activity in a minigenome assay and TRIM6 knockout cells have reduced replication of infectious EBOV, suggesting that VP35 hijacks TRIM6 to promote EBOV replication through ubiquitination. Our work provides evidence that TRIM6 is an important host cellular factor that promotes EBOV replication, and future studies will focus on whether TRIM6 could be targeted for therapeutic intervention against EBOV infection.IMPORTANCE EBOV belongs to a family of highly pathogenic viruses that cause severe hemorrhagic fever in humans and other mammals with high mortality rates (40 to 90%). Because of its high pathogenicity and lack of licensed antivirals and vaccines, EBOV is listed as a tier 1 select-agent risk group 4 pathogen. An important mechanism for the severity of EBOV infection is its suppression of innate immune responses. The EBOV VP35 protein contributes to pathogenesis, because it serves as an essential cofactor of the viral polymerase as well as a potent antagonist of innate immunity. However, how VP35 function is regulated by host cellular factors is poorly understood. Here, we report that the host E3-ubiquitin ligase TRIM6 promotes VP35 ubiquitination and is important for efficient virus replication. Therefore, our study identifies a new host factor, TRIM6, as a potential target in the development of antiviral drugs against EBOV.
Collapse
|
145
|
Liu G, Wong G, Su S, Bi Y, Plummer F, Gao GF, Kobinger G, Qiu X. Clinical Evaluation of Ebola Virus Disease Therapeutics. Trends Mol Med 2017; 23:820-830. [PMID: 28822631 DOI: 10.1016/j.molmed.2017.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 01/17/2023]
Abstract
Ebola virus disease (EVD) was first described over 40 years ago, but no treatment has been approved for humans. The 2013-2016 EVD outbreak in West Africa has expedited the clinical evaluation of several candidate therapeutics that act through different mechanisms, but with mixed results. Nevertheless, these studies are important because the accumulation of clinical data and valuable experience in conducting efficacy trials under emergency circumstances will lead to better implementation of similar studies in the future. Here, we summarize the results of EVD clinical trials, focus on the discussion of factors that may have potentially impeded the effectiveness of existing candidate therapeutics, and highlight considerations that may help meet the challenges ahead in the quest to develop clinically approved drugs.
Collapse
Affiliation(s)
- Guodong Liu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gary Wong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shuo Su
- Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuhai Bi
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Frank Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - George F Gao
- Shenzhen Key Laboratory of Pathogen and Immunity, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Gary Kobinger
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada; Département de microbiologie-infectiologie et d'immunologie, Université Laval, Québec, Canada
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
146
|
Structural Insight into Nucleoprotein Conformation Change Chaperoned by VP35 Peptide in Marburg Virus. J Virol 2017; 91:JVI.00825-17. [PMID: 28566377 DOI: 10.1128/jvi.00825-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/14/2022] Open
Abstract
Marburg virus (MARV) encodes a nucleoprotein (NP) to encapsidate its genome by oligomerization and form a ribonucleoprotein complex (RNP). According to previous investigation on nonsegmented negative-sense RNA viruses (nsNSV), the newly synthesized NPs must be prevented from indiscriminately binding to noncognate RNAs. During the viral RNA synthesis process, the RNPs undergo a transition from an RNA-bound form to a template-free form, to open access for the interaction between the viral polymerase and the RNA template. In filoviruses, this transition is regulated by VP35 peptide and other viral components. To further understand the dynamic process of filovirus RNP formation, we report here the structure of MARV NPcore, both in the apo form and in the VP35 peptide-chaperoned form. These structures reveal a typical bilobed structure, with a positive-charged RNA binding groove between two lobes. In the apo form, the MARV NP exists in an interesting hexameric state formed by the hydrophobic interaction within the long helix of the NPcore C-terminal region, which shows high structural flexibility among filoviruses and may imply critical function during RNP formation. Moreover, the VP35 peptide-chaperoned NPcore remains in a monomeric state and completely loses its affinity for single-stranded RNA (ssRNA). The structural comparison reveals that the RNA binding groove undergoes a transition from closed state to open state, chaperoned by VP35 peptide, thus preventing the interaction for viral RNA. Our investigation provides considerable structural insight into the filovirus RNP working mechanism and may support the development of antiviral therapies targeting the RNP formation of filovirus.IMPORTANCE Marburg virus is one of the most dangerous viruses, with high morbidity and mortality. A recent outbreak in Angola in 2005 caused the deaths of 272 persons. NP is one of the most essential proteins, as it encapsidates and protects the whole virus genome simultaneously with self-assembly oligomerization. Here we report the structures of MARV NPcore in two different forms. In the MARV NP apo form, we identify an interesting hexamer formed by hydrophobic interaction within a long helix, which is highly conserved and flexible among filoviruses and may indicate its critical function during the virus RNP formation. Moreover, the structural comparison with the NP-VP35 peptide complex reveals a structural transition chaperoned by VP35, in which the RNA binding groove undergoes a transition from closed state to open state. Finally, we discussed the high conservation and critical role of the VP35 binding pocket and its potential use for therapeutic development.
Collapse
|
147
|
Smith JR, Todd S, Ashander LM, Charitou T, Ma Y, Yeh S, Crozier I, Michael MZ, Appukuttan B, Williams KA, Lynn DJ, Marsh GA. Retinal Pigment Epithelial Cells are a Potential Reservoir for Ebola Virus in the Human Eye. Transl Vis Sci Technol 2017; 6:12. [PMID: 28721309 PMCID: PMC5512973 DOI: 10.1167/tvst.6.4.12] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/19/2017] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Success of Ebola virus (EBOV) as a human pathogen relates at the molecular level primarily to blockade the host cell type I interferon (IFN) antiviral response. Most individuals who survive Ebola virus disease (EVD) develop a chronic disease syndrome: approximately one-quarter of survivors suffer from uveitis, which has been associated with presence of EBOV within the eye. Clinical observations of post-Ebola uveitis indicate involvement of retinal pigment epithelial cells. METHODS We inoculated ARPE-19 human retinal pigment epithelial cells with EBOV, and followed course of infection by immunocytochemistry and measurement of titer in culture supernatant. To interrogate transcriptional responses of infected cells, we combined RNA sequencing with in silico pathway, gene ontology, transcription factor binding site, and network analyses. We measured infection-induced changes of selected transcripts by reverse transcription-quantitative polymerase chain reaction. RESULTS Human retinal pigment epithelial cells were permissive to infection with EBOV, and supported viral replication and release of virus in high titer. Unexpectedly, 28% of 560 upregulated transcripts in EBOV-infected cells were type I IFN responsive, indicating a robust type I IFN response. Following EBOV infection, cells continued to express multiple immunomodulatory molecules linked to ocular immune privilege. CONCLUSIONS Human retinal pigment epithelial cells may serve as an intraocular reservoir for EBOV, and the molecular response of infected cells may contribute to the persistence of live EBOV within the human eye. TRANSLATIONAL RELEVANCE This bedside-to-bench research links ophthalmic findings in survivors of EVD who suffer from uveitis with interactions between retinal pigment epithelial cells and EBOV.
Collapse
Affiliation(s)
- Justine R Smith
- Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia.,Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia.,South Australian Health & Medical Research Institute, SAHMRI: Mind & Brain Theme, Adelaide, Australia
| | - Shawn Todd
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Geelong, Australia
| | - Liam M Ashander
- Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia
| | - Theodosia Charitou
- South Australian Health & Medical Research Institute, SAHMRI: EMBL Australia group, Infection & Immunity Theme, Adelaide, Australia.,Systems Biology Ireland, University College Dublin, UCD Conway Institute, Dublin, Ireland
| | - Yuefang Ma
- Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia
| | - Steven Yeh
- Departments of Ophthalmology and Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ian Crozier
- Infectious Diseases Institute, Mulago Hospital Complex, Kampala, Uganda
| | - Michael Z Michael
- Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia
| | - Binoy Appukuttan
- Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia.,Flinders University School of Medicine, Eye & Vision Health, Flinders Medical Centre, Adelaide, Australia
| | - Keryn A Williams
- South Australian Health & Medical Research Institute, SAHMRI: Mind & Brain Theme, Adelaide, Australia.,Flinders University School of Medicine, Ophthalmology, Flinders Medical Centre, Adelaide, Australia
| | - David J Lynn
- Flinders University School of Medicine, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Adelaide, Australia.,South Australian Health & Medical Research Institute, SAHMRI: EMBL Australia group, Infection & Immunity Theme, Adelaide, Australia
| | - Glenn A Marsh
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Geelong, Australia
| |
Collapse
|
148
|
Dilley KA, Voorhies AA, Luthra P, Puri V, Stockwell TB, Lorenzi H, Basler CF, Shabman RS. The Ebola virus VP35 protein binds viral immunostimulatory and host RNAs identified through deep sequencing. PLoS One 2017. [PMID: 28636653 PMCID: PMC5479518 DOI: 10.1371/journal.pone.0178717] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ebola virus and Marburg virus are members of the Filovirdae family and causative agents of hemorrhagic fever with high fatality rates in humans. Filovirus virulence is partially attributed to the VP35 protein, a well-characterized inhibitor of the RIG-I-like receptor pathway that triggers the antiviral interferon (IFN) response. Prior work demonstrates the ability of VP35 to block potent RIG-I activators, such as Sendai virus (SeV), and this IFN-antagonist activity is directly correlated with its ability to bind RNA. Several structural studies demonstrate that VP35 binds short synthetic dsRNAs; yet, there are no data that identify viral immunostimulatory RNAs (isRNA) or host RNAs bound to VP35 in cells. Utilizing a SeV infection model, we demonstrate that both viral isRNA and host RNAs are bound to Ebola and Marburg VP35s in cells. By deep sequencing the purified VP35-bound RNA, we identified the SeV copy-back defective interfering (DI) RNA, previously identified as a robust RIG-I activator, as the isRNA bound by multiple filovirus VP35 proteins, including the VP35 protein from the West African outbreak strain (Makona EBOV). Moreover, RNAs isolated from a VP35 RNA-binding mutant were not immunostimulatory and did not include the SeV DI RNA. Strikingly, an analysis of host RNAs bound by wild-type, but not mutant, VP35 revealed that select host RNAs are preferentially bound by VP35 in cell culture. Taken together, these data support a model in which VP35 sequesters isRNA in virus-infected cells to avert RIG-I like receptor (RLR) activation.
Collapse
Affiliation(s)
- Kari A. Dilley
- Virology Group, J. Craig Venter Institute, Rockville, Maryland, United States of America
- * E-mail: (RSS); (KAD)
| | - Alexander A. Voorhies
- Infectious Disease Group, J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Priya Luthra
- Center for Microbial Pathogenesis, Georgia State University, Atlanta, Georgia, United States of America
| | - Vinita Puri
- Virology Group, J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Timothy B. Stockwell
- Virology Group, J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Hernan Lorenzi
- Infectious Disease Group, J. Craig Venter Institute, Rockville, Maryland, United States of America
| | - Christopher F. Basler
- Center for Microbial Pathogenesis, Georgia State University, Atlanta, Georgia, United States of America
| | - Reed S. Shabman
- Virology Group, J. Craig Venter Institute, Rockville, Maryland, United States of America
- * E-mail: (RSS); (KAD)
| |
Collapse
|
149
|
Wei H, Audet J, Wong G, He S, Huang X, Cutts T, Theriault S, Xu B, Kobinger G, Qiu X. Deep-sequencing of Marburg virus genome during sequential mouse passaging and cell-culture adaptation reveals extensive changes over time. Sci Rep 2017; 7:3390. [PMID: 28611428 PMCID: PMC5469859 DOI: 10.1038/s41598-017-03318-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/26/2017] [Indexed: 11/30/2022] Open
Abstract
Marburg virus (MARV) has caused outbreaks of filoviral hemorrhagic fever since its discovery in 1967. The largest and deadliest outbreak occurred in Angola in 2005, with 252 cases and 227 deaths. In 2014, we developed a mouse-adapted MARV, Angola variant through serial passaging in mice. The mouse-adapted MARV exhibits many of the hallmarks of MARV disease in humans. By applying deep-sequencing to every passage of the virus, we are able to study virus evolution in this host with surprising precision. We show that two regions go through substantial changes: the intergenic region between NP and VP35, as well as the first 100 amino acids of the VP40 protein. Our results also reveal that there were profound changes during the production of the final virus stock in cell culture. Overall, our results show that a handful of regions carry most of the mutations acquired during the adaptation of the virus to a new host and that many mutations become fixed very early during the adaptation process.
Collapse
Affiliation(s)
- Haiyan Wei
- Institute of Infectious Disease, Henan Center for Disease Control, Henan, China
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Jonathan Audet
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Gary Wong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Xueyong Huang
- Institute of Infectious Disease, Henan Center for Disease Control, Henan, China
| | - Todd Cutts
- Applied Biosafety Research Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Steven Theriault
- Applied Biosafety Research Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Bianli Xu
- Institute of Infectious Disease, Henan Center for Disease Control, Henan, China
| | - Gary Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Canada
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec City, Québec, Canada
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
150
|
Xu W, Luthra P, Wu C, Batra J, Leung DW, Basler CF, Amarasinghe GK. Ebola virus VP30 and nucleoprotein interactions modulate viral RNA synthesis. Nat Commun 2017; 8:15576. [PMID: 28593988 PMCID: PMC5472179 DOI: 10.1038/ncomms15576] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 04/11/2017] [Indexed: 12/20/2022] Open
Abstract
Ebola virus (EBOV) is an enveloped negative-sense RNA virus that causes sporadic outbreaks with high case fatality rates. Ebola viral protein 30 (eVP30) plays a critical role in EBOV transcription initiation at the nucleoprotein (eNP) gene, with additional roles in the replication cycle such as viral assembly. However, the mechanistic basis for how eVP30 functions during the virus replication cycle is currently unclear. Here we define a key interaction between eVP30 and a peptide derived from eNP that is important to facilitate interactions leading to the recognition of the RNA template. We present crystal structures of the eVP30 C-terminus in complex with this eNP peptide. Functional analyses of the eVP30–eNP interface identify residues that are critical for viral RNA synthesis. Altogether, these results support a model where the eVP30–eNP interaction plays a critical role in transcription initiation and provides a novel target for the development of antiviral therapy. Ebola virus (EBOV) VP30 is a multifunctional protein that plays a role in transcription, but molecular details remain unknown. Here, using X-ray crystallography and minigenome assays, Xu et al. define the interaction between VP30 and a portion of NP that is critical for optimal EBOV RNA synthesis.
Collapse
Affiliation(s)
- Wei Xu
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63105, USA
| | - Priya Luthra
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, USA
| | - Chao Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63105, USA
| | - Jyoti Batra
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63105, USA
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63105, USA
| |
Collapse
|