1
|
Kroh K, te Marvelde MR, van Greuningen LW, Laksono BM, Koopmans MPG, Kuiken T, GeurtsvanKessel CH, Embregts CWE. A comparative analysis of the dendritic cell response upon exposure to different rabies virus strains. PLoS Negl Trop Dis 2025; 19:e0012994. [PMID: 40208887 PMCID: PMC12017532 DOI: 10.1371/journal.pntd.0012994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 04/23/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025] Open
Abstract
Rabies is a viral zoonotic disease that causes over 60,000 human deaths annually worldwide. Natural infections lack a virus-specific immune response, leading to a near 100% fatality rate unless immediately treated. Rabies virus (RABV) is typically transmitted through bites from rabid dogs or other carnivores to humans and may initially interact with innate immune cells such as dendritic cells at the site of infection. This study investigates the in vitro response of human monocyte-derived dendritic cells (moDCs) exposed to two pathogenic RABV strains-silver-haired bat rabies virus (SHBRV) and dog-related rabies virus (dogRV)-and an attenuated vaccine strain (SAD P5). MoDCs were susceptible only to high doses of SHBRV and SAD P5, resulting in a more mature and migratory phenotype within the infected moDC populations. No infection was observed in moDCs exposed to dogRV. In co-culture with T cells, the presence of RABV-exposed moDCs, regardless of the strain, did not enhance T cell activation. Additionally, RABV exposure did not hinder LPS-induced moDC maturation; instead, high doses of SHBRV and SAD P5 even boosted activation levels. Overall, the findings suggest varied capabilities of RABV strains to infect and activate moDCs in vitro. However, exposure to any RABV strain did not provoke a clear antiviral state or suppression of moDC responsiveness. This lack of activation may contribute to the absence of an effective adaptive immune response in natural RABV infections.
Collapse
Affiliation(s)
- Keshia Kroh
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Brigitta M. Laksono
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Thijs Kuiken
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
2
|
Niemetz L, Bodmer BS, Olal C, Escudero-Pérez B, Hoehn K, Bencsik A, Vickers MA, Rodríguez E, Oestereich L, Hoenen T, Muñoz-Fontela C. Ebola Virus Infection of Flt3-Dependent, Conventional Dendritic Cells and Antigen Cross-presentation Leads to High Levels of T-Cell Activation. J Infect Dis 2025; 231:501-511. [PMID: 39320066 DOI: 10.1093/infdis/jiae441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Previous studies have described that Ebola virus (EBOV) infection of human monocyte-derived dendritic cells (moDCs) inhibits dendritic cell (DC) maturation, resulting in poor T-cell activation. However, it is unknown how other DC subsets distinct from moDCs respond to EBOV infection. METHODS To better understand how DCs initiate T-cell activation during EBOV infection, we assessed the response of conventional mouse DCs (cDCs) to EBOV infection utilizing a recombinant EBOV expressing the model antigen ovalbumin. RESULTS In contrast to moDCs, mouse cDC2s and cDC1s were poorly infected with EBOV but were highly activated. DCs were able to prime CD8 T cells via cross-presentation of antigens obtained from cell debris of EBOV-infected cells. EBOV infection further enhanced DC cross-presentation. CONCLUSIONS Our findings indicate that EBOV infection of cDCs results in activation rather than inhibition, leading to high levels of T-cell activation. With that we propose a mechanistic explanation for the excess T-cell activation observed in human Ebola virus disease.
Collapse
Affiliation(s)
- Linda Niemetz
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
| | - Bianca S Bodmer
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems
| | | | - Beatriz Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, Partner site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | | | | | | | | | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, Partner site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - Thomas Hoenen
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, Partner site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| |
Collapse
|
3
|
Stegeman SK, Kourko O, Amsden H, Pellizzari Delano IE, Mamatis JE, Roth M, Colpitts CC, Gee K. RNA Viruses, Toll-Like Receptors, and Cytokines: The Perfect Storm? J Innate Immun 2025; 17:126-153. [PMID: 39820070 PMCID: PMC11845175 DOI: 10.1159/000543608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs. BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs.
Collapse
Affiliation(s)
- Sophia K Stegeman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Olena Kourko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Heather Amsden
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - John E Mamatis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Madison Roth
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
4
|
Kuroda M, Halfmann PJ, Thackray LB, Diamond MS, Feldmann H, Marzi A, Kawaoka Y. An Antiviral Role for TRIM14 in Ebola Virus Infection. J Infect Dis 2023; 228:S514-S521. [PMID: 37562033 PMCID: PMC10651195 DOI: 10.1093/infdis/jiad325] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
Ebola virus (EBOV) is a highly pathogenic virus that encodes 7 multifunctional structural proteins. Multiple host factors have been reported to interact with the EBOV proteins. Here, we found that tripartite motif-containing 14 (TRIM14), an interferon-stimulated gene that mediates cellular signaling pathways associated with type I interferon and inflammatory cytokine production, interacts with EBOV nucleoprotein to enhance interferon-β (IFN-β) and nuclear factor-κB (NF-κB) promotor activation. Moreover, TRIM14 overexpression reduced viral replication in an infectious but biologically contained EBOVΔVP30 system by approximately 10-fold without affecting viral protein expression. Furthermore, TRM14-deficient mice were more susceptible to mouse-adapted EBOV infection than wild-type mice. Our data suggest that TRIM14 is a host factor with anti-EBOV activity that limits EBOV pathogenesis.
Collapse
Affiliation(s)
- Makoto Kuroda
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, Missouri, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- Pandemic Preparedness, Infection and Advanced Research Center, University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Letafati A, Salahi Ardekani O, Karami H, Soleimani M. Ebola virus disease: A narrative review. Microb Pathog 2023:106213. [PMID: 37355146 DOI: 10.1016/j.micpath.2023.106213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/23/2023] [Accepted: 06/22/2023] [Indexed: 06/26/2023]
Abstract
Ebola virus disease (EVD), which is also referred to as Ebola hemorrhagic fever, is a highly contagious and frequently lethal sickness caused by the Ebola virus. In 1976, the disease emerged in two simultaneous outbreaks in Sudan and the Democratic Republic of Congo. Subsequently, it has caused intermittent outbreaks in several African nations. The virus is primarily spread via direct contact with the bodily fluids of an infected individual or animal. EVD is distinguished by symptoms such as fever, fatigue, muscle pain, headache, and hemorrhage. The outbreak of EVD in West Africa in 2014-2016 emphasized the need for effective control and prevention measures. Despite advancements and the identification of new treatments for EVD, the primary approach to treatment continues to be centered around providing supportive care. Early detection and supportive care can enhance the likelihood of survival. This includes intravenous fluids, electrolyte replacement, and treatment of secondary infections. Experimental therapies, for instance, monoclonal antibodies and antiviral drugs, have shown promising results in animal studies and some clinical trials. Some African countries have implemented the use of vaccines developed for EVD, but their effectiveness and long-term safety are still being studied. This article provides an overview of the history, transmission, symptoms, diagnosis, treatment, epidemiology, and Ebola coinfection, as well as highlights the ongoing research efforts to develop effective treatments and vaccines to combat this deadly virus.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Salahi Ardekani
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Hassan Karami
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mina Soleimani
- Department of Laboratory Medicine, Faculty of Paramedical Sciences, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran.
| |
Collapse
|
6
|
Flórez-Álvarez L, de Souza EE, Botosso VF, de Oliveira DBL, Ho PL, Taborda CP, Palmisano G, Capurro ML, Pinho JRR, Ferreira HL, Minoprio P, Arruda E, de Souza Ferreira LC, Wrenger C, Durigon EL. Hemorrhagic fever viruses: Pathogenesis, therapeutics, and emerging and re-emerging potential. Front Microbiol 2022; 13:1040093. [PMID: 36386719 PMCID: PMC9640979 DOI: 10.3389/fmicb.2022.1040093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/06/2022] [Indexed: 01/29/2023] Open
Abstract
Hemorrhagic fever viruses (HFVs) pose a threat to global public health owing to the emergence and re-emergence of highly fatal diseases. Viral hemorrhagic fevers (VHFs) caused by these viruses are mostly characterized by an acute febrile syndrome with coagulation abnormalities and generalized hemorrhage that may lead to life-threatening organ dysfunction. Currently, the events underlying the viral pathogenicity associated with multiple organ dysfunction syndrome still underexplored. In this minireview, we address the current knowledge of the mechanisms underlying VHFs pathogenesis and discuss the available development of preventive and therapeutic options to treat these infections. Furthermore, we discuss the potential of HFVs to cause worldwide emergencies along with factors that favor their spread beyond their original niches.
Collapse
Affiliation(s)
| | | | | | | | - Paulo Lee Ho
- Virology Laboratory, Butantan Institute, São Paulo, Brazil
| | | | - Giuseppe Palmisano
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - João Renato Rebello Pinho
- Albert Einstein Institute for Teaching and Research (IIEP), Hospital Israelita Albert Einstein, São Paulo, Brazil,Hospital das Clínicas da Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Helena Lage Ferreira
- Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | | | - Eurico Arruda
- Faculty of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Luís Carlos de Souza Ferreira
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,Scientific Platform Pasteur-USP, São Paulo, Brazil
| | - Carsten Wrenger
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,*Correspondence: Carsten Wrenger, ; Edison Luiz Durigon,
| | - Edison Luiz Durigon
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,Scientific Platform Pasteur-USP, São Paulo, Brazil,*Correspondence: Carsten Wrenger, ; Edison Luiz Durigon,
| |
Collapse
|
7
|
Iba T, Levi M, Thachil J, Levy JH. Disseminated Intravascular Coagulation: The Past, Present, and Future Considerations. Semin Thromb Hemost 2022; 48:978-987. [PMID: 36100234 DOI: 10.1055/s-0042-1756300] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Disseminated intravascular coagulation (DIC) has been understood as a consumptive coagulopathy. However, impaired hemostasis is a component of DIC that occurs in a progressive manner. The critical concept of DIC is systemic activation of coagulation with vascular endothelial damage. DIC is the dynamic coagulation/fibrinolysis disorder that can proceed from compensated to decompensated phases, and is not simply impaired hemostasis, a misunderstanding that continues to evoke confusion among clinicians. DIC is a critical step of disease progression that is important to monitor over time. Impaired microcirculation and subsequent organ failure due to pathologic microthrombi formation are the pathophysiologies in sepsis-associated DIC. Impaired hemostasis due to coagulation factor depletion from hemodilution, shock, and hyperfibrinolysis occurs in trauma-associated DIC. Overt-DIC diagnostic criteria have been used clinically for more than 20 years but may not be adequate to detect the compensated phase of DIC, and due to different underlying causes, there is no "one-size-fits-all criteria." Individualized criteria for heterogeneous conditions continue to be proposed to facilitate the diagnosis. We believe that future research will provide therapeutics using new diagnostic criteria. Finally, DIC is also classified as either acute or chronic, and acute DIC results from progressive coagulation activation over a short time and requires urgent management. In this review, we examine the advances in research for DIC.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands.,Department of Medicine, Cardiometabolic Programme-NIHR UCLH/UCL BRC, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Jecko Thachil
- Department of Haematology, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
8
|
Zmrhal V, Slama P. Current knowledge about interactions between avian dendritic cells and poultry pathogens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103565. [PMID: 31830703 DOI: 10.1016/j.dci.2019.103565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 06/10/2023]
Abstract
In poultry production conditions today, birds are surrounded by viral, bacterial, and parasitic agents. DCs are the main antigen-presenting cells located in tissues of the body, and their role involves recognizing antigen structures, engulfing and processing them, and subsequently presenting antigen peptides on their surface by major histocompatibility complex, where T cells and B cells are stimulated and can begin appropriate cellular and antibody immune response. This unique function indicates that these cells can be used in producing vaccines, but first it is necessary to culture DCs in vitro to identify the principles of their interactions with pathogens. The following review summarizes our current knowledge about avian dendritic cells and their interactions with pathogens. It provides a basis for future studies of these unique cells and their use in vaccine development.
Collapse
Affiliation(s)
- Vladimir Zmrhal
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic.
| |
Collapse
|
9
|
Conservation of Structure and Immune Antagonist Functions of Filoviral VP35 Homologs Present in Microbat Genomes. Cell Rep 2020; 24:861-872.e6. [PMID: 30044983 DOI: 10.1016/j.celrep.2018.06.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 04/29/2018] [Accepted: 06/11/2018] [Indexed: 11/20/2022] Open
Abstract
Non-retroviral integrated RNA viral sequences (NIRVs) potentially encoding ∼280 amino acid homologs to filovirus VP35 proteins are present across the Myotis genus of bats. These are estimated to have been maintained for ∼18 million years, indicating their co-option. To address the reasons for co-option, 16 Myotis VP35s were characterized in comparison to VP35s from the extant filoviruses Ebola virus and Marburg virus, in which VP35s play critical roles in immune evasion and RNA synthesis. The Myotis VP35s demonstrated a conserved suppression of innate immune signaling, albeit with reduced potency, in either human or Myotis cells. Their attenuation reflects a lack of dsRNA binding that in the filoviral VP35s correlates with potent suppression of interferon responses. Despite divergent function, evolution has preserved in Myotis the structure of the filoviral VP35s, indicating that this structure is critical for co-opted function, possibly as a regulator of innate immune signaling.
Collapse
|
10
|
Edenborough KM, Bokelmann M, Lander A, Couacy-Hymann E, Lechner J, Drechsel O, Renard BY, Radonić A, Feldmann H, Kurth A, Prescott J. Dendritic Cells Generated From Mops condylurus, a Likely Filovirus Reservoir Host, Are Susceptible to and Activated by Zaire Ebolavirus Infection. Front Immunol 2019; 10:2414. [PMID: 31681302 PMCID: PMC6797855 DOI: 10.3389/fimmu.2019.02414] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/26/2019] [Indexed: 12/17/2022] Open
Abstract
Ebola virus infection of human dendritic cells (DCs) induces atypical adaptive immune responses and thereby exacerbates Ebola virus disease (EVD). Human DCs, infected with Ebola virus aberrantly express low levels of the DC activation markers CD80, CD86, and MHC class II. The T cell responses ensuing are commonly anergic rather than protective against EVD. We hypothesize that DCs derived from potential reservoir hosts such as bats, which do not develop disease signs in response to Ebola virus infection, would exhibit features associated with activation. In this study, we have examined Zaire ebolavirus (EBOV) infection of DCs derived from the Angolan free-tailed bat species, Mops condylurus. This species was previously identified as permissive to EBOV infection in vivo, in the absence of disease signs. M. condylurus has also been recently implicated as the reservoir host for Bombali ebolavirus, a virus species that is closely related to EBOV. Due to the absence of pre-existing M. condylurus species-specific reagents, we characterized its de novo assembled transcriptome and defined its phylogenetic similarity to other mammals, which enabled the identification of cross-reactive reagents for M. condylurus bone marrow-derived DC (bat-BMDC) differentiation and immune cell phenotyping. Our results reveal that bat-BMDCs are susceptible to EBOV infection as determined by detection of EBOV specific viral RNA (vRNA). vRNA increased significantly 72 h after EBOV-infection and was detected in both cells and in culture supernatants. Bat-BMDC infection was further confirmed by the observation of GFP expression in DC cultures infected with a recombinant GFP-EBOV. Bat-BMDCs upregulated CD80 and chemokine ligand 3 (CCL3) transcripts in response to EBOV infection, which positively correlated with the expression levels of EBOV vRNA. In contrast to the aberrant responses to EBOV infection that are typical for human-DC, our findings from bat-BMDCs provide evidence for an immunological basis of asymptomatic EBOV infection outcomes.
Collapse
Affiliation(s)
- Kathryn M. Edenborough
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Marcel Bokelmann
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Angelika Lander
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Emmanuel Couacy-Hymann
- LANADA, Laboratoire National d'Appui au Développement Agricole, Bingerville, Côte d'Ivoire
| | - Johanna Lechner
- Methodology and Research Infrastructure, Robert Koch Institute, Berlin, Germany
| | - Oliver Drechsel
- Methodology and Research Infrastructure, Robert Koch Institute, Berlin, Germany
| | - Bernhard Y. Renard
- Methodology and Research Infrastructure, Robert Koch Institute, Berlin, Germany
| | - Aleksandar Radonić
- Methodology and Research Infrastructure, Robert Koch Institute, Berlin, Germany
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, ON, United States
| | - Andreas Kurth
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Joseph Prescott
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
11
|
Olejnik J, Hume AJ, Leung DW, Amarasinghe GK, Basler CF, Mühlberger E. Filovirus Strategies to Escape Antiviral Responses. Curr Top Microbiol Immunol 2019; 411:293-322. [PMID: 28685291 PMCID: PMC5973841 DOI: 10.1007/82_2017_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This chapter describes the various strategies filoviruses use to escape host immune responses with a focus on innate immune and cell death pathways. Since filovirus replication can be efficiently blocked by interferon (IFN), filoviruses have evolved mechanisms to counteract both type I IFN induction and IFN response signaling pathways. Intriguingly, marburg- and ebolaviruses use different strategies to inhibit IFN signaling. This chapter also summarizes what is known about the role of IFN-stimulated genes (ISGs) in filovirus infection. These fall into three categories: those that restrict filovirus replication, those whose activation is inhibited by filoviruses, and those that have no measurable effect on viral replication. In addition to innate immunity, mammalian cells have evolved strategies to counter viral infections, including the induction of cell death and stress response pathways, and we summarize our current knowledge of how filoviruses interact with these pathways. Finally, this chapter delves into the interaction of EBOV with myeloid dendritic cells and macrophages and the associated inflammatory response, which differs dramatically between these cell types when they are infected with EBOV. In summary, we highlight the multifaceted nature of the host-viral interactions during filoviral infections.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Adam J Hume
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Christopher F Basler
- Microbial Pathogenesis, Georgia State University, Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
12
|
Muñoz-Fontela C, McElroy AK. Ebola Virus Disease in Humans: Pathophysiology and Immunity. Curr Top Microbiol Immunol 2019; 411:141-169. [PMID: 28653186 PMCID: PMC7122202 DOI: 10.1007/82_2017_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Viruses of the Ebolavirus genus cause sporadic epidemics of severe and systemic febrile disease that are fueled by human-to-human transmission. Despite the notoriety of ebolaviruses, particularly Ebola virus (EBOV), as prominent viral hemorrhagic fever agents, and the international concern regarding Ebola virus disease (EVD) outbreaks, very little is known about the pathophysiology of EVD in humans and, in particular, about the human immune correlates of survival and immune memory. This lack of basic knowledge about physiological characteristics of EVD is probably attributable to the dearth of clinical and laboratory data gathered from past outbreaks. The unprecedented magnitude of the EVD epidemic that occurred in West Africa from 2013 to 2016 has allowed, for the first time, evaluation of clinical, epidemiological, and immunological parameters in a significant number of patients using state-of-the-art laboratory equipment. This review will summarize the data from the literature regarding human pathophysiologic and immunologic responses to filoviral infection.
Collapse
Affiliation(s)
- César Muñoz-Fontela
- Laboratory of Emerging Viruses, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251, Hamburg, Germany.
| | - Anita K McElroy
- Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
13
|
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
|
14
|
Supramaniam A, Lui H, Bellette BM, Rudd PA, Herrero LJ. How myeloid cells contribute to the pathogenesis of prominent emerging zoonotic diseases. J Gen Virol 2018; 99:953-969. [DOI: 10.1099/jgv.0.001024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Aroon Supramaniam
- 1Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | - Hayman Lui
- 2School of Medicine, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | | | - Penny A. Rudd
- 1Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | - Lara J. Herrero
- 1Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
- 2School of Medicine, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| |
Collapse
|
15
|
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
|
16
|
Dutta P, Halder AK, Basu S, Kundu M. A survey on Ebola genome and current trends in computational research on the Ebola virus. Brief Funct Genomics 2017; 17:374-380. [DOI: 10.1093/bfgp/elx020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
17
|
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: 69] [Impact Index Per Article: 8.6] [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
|
18
|
|
19
|
Basler CF. Molecular pathogenesis of viral hemorrhagic fever. Semin Immunopathol 2017; 39:551-561. [PMID: 28555386 DOI: 10.1007/s00281-017-0637-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/23/2017] [Indexed: 12/11/2022]
Abstract
The clinical syndrome referred to as viral hemorrhagic fever (VHF) can be caused by several different families of RNA viruses, including select members of the arenaviruses, bunyaviruses, filoviruses, and flaviviruses. VHF is characterized by malaise, fever, vascular permeability, decreased plasma volume, coagulation abnormalities, and varying degrees of hemorrhage. Study of the filovirus Ebola virus has demonstrated a critical role for suppression of innate antiviral defenses in viral pathogenesis. Additionally, antigen-presenting cells are targets of productive infection and immune dysregulation. Among these cell populations, monocytes and macrophages are proposed to produce damaging inflammatory cytokines, while infected dendritic cells fail to undergo proper maturation, potentially impairing adaptive immunity. Uncontrolled virus replication and accompanying inflammatory responses are thought to promote vascular leakage and coagulopathy. However, the specific molecular pathways that underlie these features of VHF remain poorly understood. The arenavirus Lassa virus and the flavivirus yellow fever virus exhibit similar molecular pathogenesis suggesting common underlying mechanisms. Because non-human primate models that closely mimic VHF are available for Ebola, Lassa, and yellow fever viruses, we propose that comparative molecular studies using these models will yield new insights into the molecular underpinnings of VHF and suggest new therapeutic approaches.
Collapse
Affiliation(s)
- Christopher F Basler
- Center for Microbial Pathogenesis, Georgia Research Alliance Eminent Scholar in Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
20
|
Abstract
INTRODUCTION The potential roles of toll-like receptors (TLRs) in immunopathogenesis of Ebola virus disease should be unraveled to provoke possible prophylactic or therapeutic implications of TLRs for EVD. Areas covered: The Ebola virus (EBOV) infection virtually paralyses all the main mechanisms responsible for induction of type I interferon (IFN-I) response. To summarize, EBOV infection interferes with: a) the TIR-domain-containing adapter-inducing interferon-β (TRIF) pathway that is mediated by TLR3 and TLR4 signaling; b) the interferon regulatory factor 7 (IRF7) pathway that is stimulated by TLR7 and TLR9; c) the intracellular signaling that is induced by retinoic acid-inducible gene 1 (RIG-I)-like receptors (RLRs); and d) the autocrine/paracrine feedback loop that is mediated by the IFN-stimulated gene factor 3 (ISGF3) complex. Upon infection with EBOV infection, TLR4 plays a key role in production of proinflammatory mediators. Expert opinion: It is theoretically possible that use of TLRs 3, 4, 7, and 9 agonists would be beneficial to improve the IFN-I response, despite their systemic side effects. Also, antagonist of TLR4 can be utilized to prevent production of proinflammatory cytokines. Additionally, it is highly recommended to design future investigations aimed at determining if the utilization of IFN-I would be beneficial for prophylactic/therapeutic programs of Ebola.
Collapse
Affiliation(s)
- Amene Saghazadeh
- a Research Center for Immunodeficiencies, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran.,b Systematic Review and Meta-Analysis Expert Group (SRMEG) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - Nima Rezaei
- a Research Center for Immunodeficiencies, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran.,b Systematic Review and Meta-Analysis Expert Group (SRMEG) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran.,c Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,d Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA) , Universal Scientific Education and Research Network (USERN) , Sheffield , UK
| |
Collapse
|
21
|
Baseler L, Chertow DS, Johnson KM, Feldmann H, Morens DM. The Pathogenesis of Ebola Virus Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:387-418. [DOI: 10.1146/annurev-pathol-052016-100506] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Laura Baseler
- Department of Veterinary Medicine and Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Daniel S. Chertow
- Critical Care Medicine Department, Clinical Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Karl M. Johnson
- Founder, Special Pathogens Branch, Centers for Disease Control and Prevention, Placitas, New Mexico 87043
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840
| | - David M. Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892;
| |
Collapse
|
22
|
Forsyth KS, Eisenlohr LC. Giving CD4+ T cells the slip: viral interference with MHC class II-restricted antigen processing and presentation. Curr Opin Immunol 2016; 40:123-9. [PMID: 27115617 PMCID: PMC4894315 DOI: 10.1016/j.coi.2016.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 03/08/2016] [Indexed: 01/18/2023]
Abstract
Activation of CD4+ T cells through interactions with peptides bound to Major Histocompatibility Complex Class II (MHC-II) molecules is a crucial step in clearance of most pathogens. Consequently, many viruses have evolved ways of blocking this aspect of adaptive immunity, from specific targeting of processing and presentation components to modulation of signaling pathways that regulate peptide presentation in addition to many other host defense mechanisms. Such cases of interference are far less common compared to what has been elucidated in MHC-I processing and presentation. This may be attributable in part to the complexity of MHC-II antigen processing, the scope of which is only now coming to light.
Collapse
Affiliation(s)
- Katherine S Forsyth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Laurence C Eisenlohr
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Pathology and Laboratory Medicine at the Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Effects of Filovirus Interferon Antagonists on Responses of Human Monocyte-Derived Dendritic Cells to RNA Virus Infection. J Virol 2016; 90:5108-5118. [PMID: 26962215 DOI: 10.1128/jvi.00191-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/06/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Dendritic cells (DCs) are major targets of filovirus infection in vivo Previous studies have shown that the filoviruses Ebola virus (EBOV) and Marburg virus (MARV) suppress DC maturation in vitro Both viruses also encode innate immune evasion functions. The EBOV VP35 (eVP35) and the MARV VP35 (mVP35) proteins each can block RIG-I-like receptor signaling and alpha/beta interferon (IFN-α/β) production. The EBOV VP24 (eVP24) and MARV VP40 (mVP40) proteins each inhibit the production of IFN-stimulated genes (ISGs) by blocking Jak-STAT signaling; however, this occurs by different mechanisms, with eVP24 blocking nuclear import of tyrosine-phosphorylated STAT1 and mVP40 blocking Jak1 function. MARV VP24 (mVP24) has been demonstrated to modulate host cell antioxidant responses. Previous studies demonstrated that eVP35 is sufficient to strongly impair primary human monocyte-derived DC (MDDC) responses upon stimulation induced through the RIG-I-like receptor pathways. We demonstrate that mVP35, like eVP35, suppresses not only IFN-α/β production but also proinflammatory responses after stimulation of MDDCs with RIG-I activators. In contrast, eVP24 and mVP40, despite suppressing ISG production upon RIG-I activation, failed to block upregulation of maturation markers or T cell activation. mVP24, although able to stimulate expression of antioxidant response genes, had no measurable impact of DC function. These data are consistent with a model where filoviral VP35 proteins are the major suppressors of DC maturation during filovirus infection, whereas the filoviral VP24 proteins and mVP40 are insufficient to prevent DC maturation. IMPORTANCE The ability to suppress the function of dendritic cells (DCs) likely contributes to the pathogenesis of disease caused by the filoviruses Ebola virus and Marburg virus. To clarify the basis for this DC suppression, we assessed the effect of filovirus proteins known to antagonize innate immune signaling pathways, including Ebola virus VP35 and VP24 and Marburg virus VP35, VP40, and VP24, on DC maturation and function. The data demonstrate that the VP35s from Ebola virus and Marburg virus are the major suppressors of DC maturation and that the effects on DCs of the remaining innate immune inhibitors are minor.
Collapse
|
24
|
Messaoudi I, Amarasinghe GK, Basler CF. Filovirus pathogenesis and immune evasion: insights from Ebola virus and Marburg virus. Nat Rev Microbiol 2015; 13:663-76. [PMID: 26439085 DOI: 10.1038/nrmicro3524] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ebola viruses and Marburg viruses, members of the filovirus family, are zoonotic pathogens that cause severe disease in people, as highlighted by the latest Ebola virus epidemic in West Africa. Filovirus disease is characterized by uncontrolled virus replication and the activation of host responses that contribute to pathogenesis. Underlying these phenomena is the potent suppression of host innate antiviral responses, particularly the type I interferon response, by viral proteins, which allows high levels of viral replication. In this Review, we describe the mechanisms used by filoviruses to block host innate immunity and discuss the links between immune evasion and filovirus pathogenesis.
Collapse
Affiliation(s)
- Ilhem Messaoudi
- School of Medicine, University of California Riverside, Riverside, California 92521, USA
| | - Gaya K Amarasinghe
- The Division of Biology &Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA
| | - Christopher F Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
25
|
Raj U, Varadwaj PK. Flavonoids as Multi-target Inhibitors for Proteins Associated with Ebola Virus: In Silico Discovery Using Virtual Screening and Molecular Docking Studies. Interdiscip Sci 2015; 8:132-141. [PMID: 26286008 DOI: 10.1007/s12539-015-0109-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/15/2014] [Accepted: 01/14/2015] [Indexed: 10/23/2022]
Abstract
Ebola virus is a single-stranded, negative-sense RNA virus that causes severe hemorrhagic fever in humans and non-human primates. This virus is unreceptive to a large portion of the known antiviral drugs, and there is no valid treatment as on date for disease created by this pathogen. Looking into its ability to create a pandemic scenario across globe, there is an utmost need for new drugs and therapy to combat this life-threatening infection. The current study deals with the evaluation of the inhibitory activity of flavonoids against the four selected Ebola virus receptor proteins, using in silico studies. The viral proteins VP40, VP35, VP30 and VP24 were docked with small molecules obtained from flavonoid class and its derivatives and evaluated on the basis of energetics, stereochemical considerations and pharmacokinetic properties to identify potential lead compounds. The results showed that both top-ranking screened flavonoids, i.e., Gossypetin and Taxifolin, showed better docking scores and binding energies in all the EBOV receptors when compared to those of the reported compound. All the screened flavonoids have known antiviral activity, acceptable pharmacokinetic properties and are being used on human and thus can be taken as anti-Ebola therapy without the time lag for clinical trial.
Collapse
Affiliation(s)
- Utkarsh Raj
- Bioinformatics Division, Indian Institute of Information Technology, Allahabad, India.
| | | |
Collapse
|
26
|
Messaoudi I, Basler CF. Immunological features underlying viral hemorrhagic fevers. Curr Opin Immunol 2015; 36:38-46. [PMID: 26163194 DOI: 10.1016/j.coi.2015.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 11/28/2022]
Abstract
Several enveloped RNA viruses of the arenavirus, bunyavirus, filovirus and flavivirus families are associated with a syndrome known as viral hemorrhagic fever (VHF). VHF is characterized by fever, vascular leakage, coagulation defects and multi organ system failure. VHF is currently viewed as a disease precipitated by viral suppression of innate immunity, which promotes systemic virus replication and excessive proinflammatory cytokine responses that trigger the manifestations of severe disease. However, the mechanisms by which immune dysregulation contributes to disease remain poorly understood. Infection of nonhuman primates closely recapitulates human VHF, notably Ebola and yellow fever, thereby providing excellent models to better define the immunological basis for this syndrome. Here we review the current state of our knowledge and suggest future directions that will better define the immunological mechanisms underlying VHF.
Collapse
Affiliation(s)
- Ilhem Messaoudi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States
| | - Christopher F Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
27
|
Basler CF. Innate immune evasion by filoviruses. Virology 2015; 479-480:122-30. [DOI: 10.1016/j.virol.2015.03.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/17/2015] [Indexed: 01/07/2023]
|
28
|
Rougeron V, Feldmann H, Grard G, Becker S, Leroy EM. Ebola and Marburg haemorrhagic fever. J Clin Virol 2015; 64:111-9. [PMID: 25660265 PMCID: PMC11080958 DOI: 10.1016/j.jcv.2015.01.014] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 01/15/2015] [Accepted: 01/16/2015] [Indexed: 01/02/2023]
Abstract
Ebolaviruses and Marburgviruses (family Filoviridae) are among the most virulent pathogens for humans and great apes causing severe haemorrhagic fever and death within a matter of days. This group of viruses is characterized by a linear, non-segmented, single-stranded RNA genome of negative polarity. The overall burden of filovirus infections is minimal and negligible compared to the devastation caused by malnutrition and other infectious diseases prevalent in Africa such as malaria, dengue or tuberculosis. In this paper, we review the knowledge gained on the eco/epidemiology, the pathogenesis and the disease control measures for Marburg and Ebola viruses developed over the last 15 years. The overall progress is promising given the little attention that these pathogen have achieved in the past; however, more is to come over the next decade given the more recent interest in these pathogens as potential public and animal health concerns. Licensing of therapeutic and prophylactic options may be achievable over the next 5-10 years.
Collapse
Affiliation(s)
- V Rougeron
- Laboratoire MiVEGEC, UMR IRD 224 CNRS 5290 UMI, 911 Av. Agropolis, 34394 Montpellier, Cedex 5, France; International Center for Medical Research of Franceville, BP769, Franceville, Gabon.
| | - H Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - G Grard
- International Center for Medical Research of Franceville, BP769, Franceville, Gabon
| | - S Becker
- Institut für Virologie, Philipps-Universität Marburg, Hans-Meerwein-Str. 2, Marburg 35037, Germany
| | - E M Leroy
- Laboratoire MiVEGEC, UMR IRD 224 CNRS 5290 UMI, 911 Av. Agropolis, 34394 Montpellier, Cedex 5, France; International Center for Medical Research of Franceville, BP769, Franceville, Gabon
| |
Collapse
|
29
|
Yen B, Mulder LCF, Martinez O, Basler CF. Molecular basis for ebolavirus VP35 suppression of human dendritic cell maturation. J Virol 2014; 88:12500-10. [PMID: 25142601 PMCID: PMC4248944 DOI: 10.1128/jvi.02163-14] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/10/2014] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Zaire ebolavirus (EBOV) VP35 is a double-stranded RNA (dsRNA)-binding protein that inhibits RIG-I signaling and alpha/beta interferon (IFN-α/β) responses by both dsRNA-binding-dependent and -independent mechanisms. VP35 also suppresses dendritic cell (DC) maturation. Here, we define the pathways and mechanisms through which VP35 impairs DC maturation. Wild-type VP35 (VP35-WT) and two well-characterized VP35 mutants (F239A and R322A) that independently ablate dsRNA binding and RIG-I inhibition were delivered to primary human monocyte-derived DCs (MDDCs) using a lentivirus-based expression system. VP35-WT suppressed not only IFN-α/β but also proinflammatory responses following stimulation of MDDCs with activators of RIG-I-like receptor (RLR) signaling, including RIG-I activators such as Sendai virus (SeV) or 5'-triphosphate RNA, or MDA5 activators such as encephalomyocarditis virus (EMCV) or poly(I · C). The F239A and R322A mutants exhibited greatly reduced suppression of IFN-α/β and proinflammatory cytokine production following treatment of DCs with RLR agonists. VP35-WT also blocked the upregulation of DC maturation markers and the stimulation of allogeneic T cell responses upon SeV infection, whereas the mutants did not. In contrast to the RLR activators, VP35-WT and the VP35 mutants impaired IFN-β production induced by Toll-like receptor 3 (TLR3) or TLR4 agonists but failed to inhibit proinflammatory cytokine production induced by TLR2, TLR3, or TLR4 agonists. Furthermore, VP35 did not prevent lipopolysaccharide (LPS)-induced upregulation of surface markers of MDDC maturation and did not prevent LPS-triggered allogeneic T cell stimulation. Therefore, VP35 is a general antagonist of DC responses to RLR activation. However, TLR agonists can circumvent many of the inhibitory effects of VP35. Therefore, it may be possible to counteract EBOV immune evasion by using treatments that bypass the VP35-imposed block to DC maturation. IMPORTANCE The VP35 protein, which is an inhibitor of RIG-I signaling and alpha/beta interferon (IFN-α/β) responses, has been implicated as an EBOV-encoded factor that contributes to suppression of dendritic cell (DC) function. We used wild-type VP35 and previously characterized VP35 mutants to clarify VP35-DC interactions. Our data demonstrate that VP35 is a general inhibitor of RIG-I-like receptor (RLR) signaling that blocks not only RIG-I- but also MDA5-mediated induction of IFN-α/β responses. Furthermore, in DCs, VP35 also impairs the RLR-mediated induction of proinflammatory cytokine production, upregulation of costimulatory markers, and activation of T cells. These inhibitory activities require VP35 dsRNA-binding activity, an activity previously correlated to VP35 RIG-I inhibitory function. In contrast, while VP35 can inhibit IFN-α/β production induced by TLR3 or TLR4 agonists, this occurs in a dsRNA-independent fashion, and VP35 does not inhibit TLR-mediated expression of proinflammatory cytokines. These data suggest strategies to overcome VP35 inhibition of DC function.
Collapse
Affiliation(s)
- Benjamin Yen
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lubbertus C F Mulder
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Osvaldo Martinez
- Department of Biology, Winona State University, Winona, Minnesota, USA
| | - Christopher F Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
30
|
Wong G, Kobinger GP, Qiu X. Characterization of host immune responses in Ebola virus infections. Expert Rev Clin Immunol 2014; 10:781-90. [PMID: 24742338 DOI: 10.1586/1744666x.2014.908705] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ebola causes highly lethal hemorrhagic fever in humans with no licensed countermeasures. Its virulence can be attributed to several immunoevasion mechanisms: an early inhibition of innate immunity started by the downregulation of type I interferon, epitope masking and subversion of the adaptive humoural immunity by secreting a truncated form of the viral glycoprotein. Deficiencies in specific and non-specific antiviral responses result in unrestricted viral replication and dissemination in the host, causing death typically within 10 days after the appearance of symptoms. This review summarizes the host immune response to Ebola infection, and highlights the short- and long-term immune responses crucial for protection, which holds implications for the design of future vaccines and therapeutics.
Collapse
Affiliation(s)
- Gary Wong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street Winnipeg, MB, R3E 3R2 Canada
| | | | | |
Collapse
|
31
|
Abstract
Zoonotic transmission of Ebola virus (EBOV) to humans causes a severe haemorrhagic fever in afflicted individuals with high case-fatality rates. Neither vaccines nor therapeutics are at present available to combat EBOV infection, making the virus a potential threat to public health. To devise antiviral strategies, it is important to understand which components of the immune system could be effective against EBOV infection. The interferon (IFN) system constitutes a key innate defence against viral infections and prevents development of lethal disease in mice infected with EBOV strains not adapted to this host. Recent research revealed that expression of the host cell IFN-inducible transmembrane proteins 1-3 (IFITM1-3) and tetherin is induced by IFN and restricts EBOV infection, at least in cell culture model systems. IFITMs, tetherin and other effector molecules of the IFN system could thus pose a potent barrier against EBOV spread in humans. However, EBOV interferes with signalling events required for human cells to express these proteins. Here, we will review the strategies employed by EBOV to fight the IFN system, and we will discuss how IFITM proteins and tetherin inhibit EBOV infection.
Collapse
Affiliation(s)
- A Kühl
- Institute of Virology, Hannover Medical School, Hannover, Germany.
| | | |
Collapse
|
32
|
Lubaki NM, Ilinykh P, Pietzsch C, Tigabu B, Freiberg AN, Koup RA, Bukreyev A. The lack of maturation of Ebola virus-infected dendritic cells results from the cooperative effect of at least two viral domains. J Virol 2013; 87:7471-7485. [PMID: 23616668 PMCID: PMC3700277 DOI: 10.1128/jvi.03316-12] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 04/17/2013] [Indexed: 11/20/2022] Open
Abstract
Ebola virus (EBOV) infections are characterized by deficient T lymphocyte responses, T lymphocyte apoptosis, and lymphopenia in the absence of direct infection of T lymphocytes. In contrast, dendritic cells (DC) are infected but fail to mature appropriately, thereby impairing the T cell response. We investigated the contributions of EBOV proteins in modulating DC maturation by generating recombinant viruses expressing enhanced green fluorescent protein and carrying mutations affecting several potentially immunomodulating domains. They included envelope glycoprotein (GP) domains, as well as innate response antagonist domains (IRADs) previously identified in the VP24 and VP35 proteins. GP expressed by an unrelated vector, but not the wild-type EBOV, was found to strongly induce DC maturation, and infections with recombinant EBOV carrying mutations disabling GP functional domains did not restore DC maturation. In contrast, each of the viruses carrying mutations disabling any IRAD in VP35 induced a dramatic upregulation of DC maturation markers. This was dependent on infection, but not interaction with GP. Disabling of IRADs also resulted in up to a several hundredfold increase in secretion of cytokines and chemokines. Furthermore, these mutations induced formation of homotypic DC clusters, which represent close correlates of their maturation and presumably facilitate transfer of antigen from migratory DC to lymph node DC. Thus, an individual IRAD is insufficient to suppress DC maturation; rather, the suppression of DC maturation and the "immune paralysis" observed during EBOV infections results from a cooperative effect of two or more individual IRADs.
Collapse
Affiliation(s)
- Ndongala M. Lubaki
- Departments of Pathology
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Philipp Ilinykh
- Departments of Pathology
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Colette Pietzsch
- Departments of Pathology
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bersabeh Tigabu
- Departments of Pathology
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Alexander N. Freiberg
- Departments of Pathology
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard A. Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander Bukreyev
- Departments of Pathology
- Microbiology and Immunology
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
33
|
Mattoscio D, Segré CV, Chiocca S. Viral manipulation of cellular protein conjugation pathways: The SUMO lesson. World J Virol 2013; 2:79-90. [PMID: 24175232 PMCID: PMC3785051 DOI: 10.5501/wjv.v2.i2.79] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/23/2013] [Accepted: 02/06/2013] [Indexed: 02/05/2023] Open
Abstract
Small ubiquitin-like modifier (SUMO)ylation is a key post-translational modification mechanism that controls the function of a plethora of proteins and biological processes. Given its central regulatory role, it is not surprising that it is widely exploited by viruses. A number of viral proteins are known to modify and/or be modified by the SUMOylation system to exert their function, to create a cellular environment more favorable for virus survival and propagation, and to prevent host antiviral responses. Since the SUMO pathway is a multi-step cascade, viral proteins engage with it at many levels, to advance and favor each stage of a typical infection cycle: replication, viral assembly and immune evasion. Here we review the current knowledge on the interplay between the host SUMO system and viral lifecycle.
Collapse
|
34
|
Abstract
Since viruses rely on functional cellular machinery for efficient propagation, apoptosis is an important mechanism to fight viral infections. In this study, we sought to determine the mechanism of cell death caused by Ebola virus (EBOV) infection by assaying for multiple stages of apoptosis and hallmarks of necrosis. Our data indicate that EBOV does not induce apoptosis in infected cells but rather leads to a nonapoptotic form of cell death. Ultrastructural analysis confirmed necrotic cell death of EBOV-infected cells. To investigate if EBOV blocks the induction of apoptosis, infected cells were treated with different apoptosis-inducing agents. Surprisingly, EBOV-infected cells remained sensitive to apoptosis induced by external stimuli. Neither receptor- nor mitochondrion-mediated apoptosis signaling was inhibited in EBOV infection. Although double-stranded RNA (dsRNA)-induced activation of protein kinase R (PKR) was blocked in EBOV-infected cells, induction of apoptosis mediated by dsRNA was not suppressed. When EBOV-infected cells were treated with dsRNA-dependent caspase recruiter (dsCARE), an antiviral protein that selectively induces apoptosis in cells containing dsRNA, virus titers were strongly reduced. These data show that the inability of EBOV to block apoptotic pathways may open up new strategies toward the development of antiviral therapeutics.
Collapse
|
35
|
Sullivan NJ, Fausther-Bovendo H, Nabel GJ. Ebola vaccine. Vaccines (Basel) 2013. [DOI: 10.1016/b978-1-4557-0090-5.00049-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
36
|
Paessler S, Walker DH. Pathogenesis of the viral hemorrhagic fevers. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:411-40. [PMID: 23121052 DOI: 10.1146/annurev-pathol-020712-164041] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Four families of enveloped RNA viruses, filoviruses, flaviviruses, arenaviruses, and bunyaviruses, cause hemorrhagic fevers. These viruses are maintained in specific natural cycles involving nonhuman primates, bats, rodents, domestic ruminants, humans, mosquitoes, and ticks. Vascular instability varies from mild to fatal shock, and hemorrhage ranges from none to life threatening. The pathogenic mechanisms are extremely diverse and include deficiency of hepatic synthesis of coagulation factors owing to hepatocellular necrosis, cytokine storm, increased permeability by vascular endothelial growth factor, complement activation, and disseminated intravascular coagulation in one or more hemorrhagic fevers. The severity of disease caused by these agents varies tremendously; there are extremely high fatality rates in Ebola and Marburg hemorrhagic fevers, and asymptomatic infection predominates in yellow fever and dengue viral infections. Although ineffective immunity and high viral loads are characteristic of several viral hemorrhagic fevers, severe plasma leakage occurs at the time of viral clearance and defervescence in dengue hemorrhagic fever.
Collapse
Affiliation(s)
- Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | | |
Collapse
|
37
|
Abstract
Marburg and Ebola viruses cause a severe hemorrhagic disease in humans with high fatality rates. Early target cells of filoviruses are monocytes, macrophages, and dendritic cells. The infection spreads to the liver, spleen and later other organs by blood and lymph flow. A hallmark of filovirus infection is the depletion of non-infected lymphocytes; however, the molecular mechanisms leading to the observed bystander lymphocyte apoptosis are poorly understood. Also, there is limited knowledge about the fate of infected cells in filovirus disease. In this review we will explore what is known about the intracellular events leading to virus amplification and cell damage in filovirus infection. Furthermore, we will discuss how cellular dysfunction and cell death may correlate with disease pathogenesis.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; E-Mails: (J.O.); (R.B.C.)
- National Emerging Infectious Diseases Laboratories Institute, Boston University, 72 East Concord Street, Boston, MA 02118, USA
| | - Elena Ryabchikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Science, Pr. Lavrent’eva, 8, Novosibirsk 630090, Russian Federation; E-Mail:
| | - Ronald B. Corley
- Department of Microbiology, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; E-Mails: (J.O.); (R.B.C.)
- National Emerging Infectious Diseases Laboratories Institute, Boston University, 72 East Concord Street, Boston, MA 02118, USA
| | - Elke Mühlberger
- Department of Microbiology, School of Medicine, Boston University, 72 East Concord Street, Boston, MA 02118, USA; E-Mails: (J.O.); (R.B.C.)
- National Emerging Infectious Diseases Laboratories Institute, Boston University, 72 East Concord Street, Boston, MA 02118, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-617-638-0336; Fax: +1-617-638-4286
| |
Collapse
|
38
|
Martinez O, Leung LW, Basler CF. The role of antigen-presenting cells in filoviral hemorrhagic fever: gaps in current knowledge. Antiviral Res 2012; 93:416-28. [PMID: 22333482 PMCID: PMC3299938 DOI: 10.1016/j.antiviral.2012.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 01/26/2012] [Accepted: 01/30/2012] [Indexed: 12/01/2022]
Abstract
The filoviruses, Ebola virus (EBOV) and Marburg virus (MARV), are highly lethal zoonotic agents of concern as emerging pathogens and potential bioweapons. Antigen-presenting cells (APCs), particularly macrophages and dendritic cells, are targets of filovirus infection in vivo. Infection of these cell types has been proposed to contribute to the inflammation, activation of coagulation cascades and ineffective immune responses characteristic of filovirus hemorrhagic fever. However, many aspects of filovirus–APC interactions remain to be clarified. Among the unanswered questions: What determines the ability of filoviruses to replicate in different APC subsets? What are the cellular signaling pathways that sense infection and lead to production of copious quantities of cytokines, chemokines and tissue factor? What are the mechanisms by which innate antiviral responses are disabled by these viruses, and how may these mechanisms contribute to inadequate adaptive immunity? A better understanding of these issues will clarify the pathogenesis of filoviral hemorrhagic fever and provide new avenues for development of therapeutics.
Collapse
Affiliation(s)
- Osvaldo Martinez
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
39
|
dsRNA binding characterization of full length recombinant wild type and mutants Zaire ebolavirus VP35. Antiviral Res 2012; 93:354-63. [PMID: 22289166 PMCID: PMC7114247 DOI: 10.1016/j.antiviral.2012.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 12/16/2011] [Accepted: 01/15/2012] [Indexed: 12/13/2022]
Abstract
The Ebola viruses (EBOVs) VP35 protein is a multifunctional major virulence factor involved in EBOVs replication and evasion of the host immune system. EBOV VP35 is an essential component of the viral RNA polymerase, it is a key participant of the nucleocapsid assembly and it inhibits the innate immune response by antagonizing RIG-I like receptors through its dsRNA binding function and, hence, by suppressing the host type I interferon (IFN) production. Insights into the VP35 dsRNA recognition have been recently revealed by structural and functional analysis performed on its C-terminus protein. We report the biochemical characterization of the Zaire ebolavirus (ZEBOV) full-length recombinant VP35 (rVP35)–dsRNA binding function. We established a novel in vitro magnetic dsRNA binding pull down assay, determined the rVP35 optimal dsRNA binding parameters, measured the rVP35 equilibrium dissociation constant for heterologous in vitro transcribed dsRNA of different length and short synthetic dsRNA of 8 bp, and validated the assay for compound screening by assessing the inhibitory ability of auryntricarboxylic acid (IC50 value of 50 μg/mL). Furthermore, we compared the dsRNA binding properties of full length wt rVP35 with those of R305A, K309A and R312A rVP35 mutants, which were previously reported to be defective in dsRNA binding-mediated IFN inhibition, showing that the latter have measurably increased Kd values for dsRNA binding and modified migration patterns in mobility shift assays with respect to wt rVP35. Overall, these results provide the first characterization of the full-length wt and mutants VP35–dsRNA binding functions.
Collapse
|
40
|
Ebihara H, Rockx B, Marzi A, Feldmann F, Haddock E, Brining D, LaCasse RA, Gardner D, Feldmann H. Host response dynamics following lethal infection of rhesus macaques with Zaire ebolavirus. J Infect Dis 2011; 204 Suppl 3:S991-9. [PMID: 21987781 DOI: 10.1093/infdis/jir336] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To gain further insight into the interdependent pathogenic processes in Ebola hemorrhagic fever (EHF), we have examined the dynamics of host responses in individual rhesus macaques infected with Zaire ebolavirus over the entire disease course. Examination of coagulation parameters revealed that decreased coagulation inhibitor activity triggered severe coagulopathy as indicated by prolonged coagulation times and decreased fibrinogen levels. This has been proposed as one of the significant mechanisms underlying disseminated intravascular coagulation in EHF patients. Furthermore, monitoring of expression levels for cytokines/chemokines suggested a mixed anti-inflammatory response syndrome (MARS), which indicates that a catastrophic uncontrolled immunological status contributes to the development of fatal hemorrhagic fever. These results highlight the pathological analogies between EHF and severe sepsis and not only contribute to our understanding of the pathogenic process, but will also help to establish novel postexposure treatment modalities.
Collapse
Affiliation(s)
- Hideki Ebihara
- Laboratory of Virology, Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana 59840, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Leroy EM, Gonzalez JP, Baize S. Ebola and Marburg haemorrhagic fever viruses: major scientific advances, but a relatively minor public health threat for Africa. Clin Microbiol Infect 2011; 17:964-76. [PMID: 21722250 DOI: 10.1111/j.1469-0691.2011.03535.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ebola and Marburg viruses are the only members of the Filoviridae family (order Mononegavirales), a group of viruses characterized by a linear, non-segmented, single-strand negative RNA genome. They are among the most virulent pathogens for humans and great apes, causing acute haemorrhagic fever and death within a matter of days. Since their discovery 50 years ago, filoviruses have caused only a few outbreaks, with 2317 clinical cases and 1671 confirmed deaths, which is negligible compared with the devastation caused by malnutrition and other infectious diseases prevalent in Africa (malaria, cholera, AIDS, dengue, tuberculosis …). Yet considerable human and financial resourses have been devoted to research on these viruses during the past two decades, partly because of their potential use as bioweapons. As a result, our understanding of the ecology, host interactions, and control of these viruses has improved considerably.
Collapse
Affiliation(s)
- E M Leroy
- Centre International de Recherches Médicales de Franceville, Franceville, Gabon.
| | | | | |
Collapse
|
42
|
Leung LW, Park MS, Martinez O, Valmas C, López CB, Basler CF. Ebolavirus VP35 suppresses IFN production from conventional but not plasmacytoid dendritic cells. Immunol Cell Biol 2011; 89:792-802. [PMID: 21263462 PMCID: PMC4148147 DOI: 10.1038/icb.2010.169] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Ebolaviruses naturally infect a wide variety of cells including macrophages and dendritic cells (DCs), and the resulting cytokine and interferon-α/β (IFN) responses of infected cells are thought to influence viral pathogenesis. The VP35 protein impairs RIG-I-like receptor-dependent signaling to inhibit IFN production, and this function has been suggested to promote the ineffective host immune response characteristic of ebolavirus infection. To assess the impact of VP35 on innate immunity in biologically relevant primary cells, we used a recombinant Newcastle disease virus encoding VP35 (NDV/VP35) to infect macrophages and conventional DCs, which primarily respond to RNA virus infection via RIG-I-like pathways. VP35 suppressed not only IFN but also tumor necrosis factor (TNF)-α secretion, which are normally produced from these cells upon NDV infection. Additionally, in cells susceptible to the activity of VP35, IRF7 activation is impaired. In contrast, NDV/VP35 infection of plasmacytoid DCs, which activate IRF7 and produce IFN through TLR-dependent signaling, leads to robust IFN production. When plasmacytoid DCs deficient for TLR signaling were infected, NDV/VP35 was able to inhibit IFN production. Consistent with this, VP35 was less able to inhibit TLR-dependent versus RIG-I-dependent signaling in vitro. These data demonstrate that ebolavirus VP35 suppresses both IFN and cytokine production in multiple primary human cell types. However, cells that utilize the TLR pathway can circumvent this inhibition, suggesting that the presence of multiple viral sensors enables the host to overcome viral immune evasion mechanisms.
Collapse
Affiliation(s)
- Lawrence W. Leung
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Man-Seong Park
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Osvaldo Martinez
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Charalampos Valmas
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Carolina B. López
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| | - Christopher F. Basler
- Department of Microbiology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029
| |
Collapse
|
43
|
Bradfute SB, Bavari S. Correlates of immunity to filovirus infection. Viruses 2011; 3:982-1000. [PMID: 21994766 PMCID: PMC3185794 DOI: 10.3390/v3070982] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 06/14/2011] [Accepted: 06/16/2011] [Indexed: 12/13/2022] Open
Abstract
Filoviruses can cause severe, often fatal hemorrhagic fever in humans. Recent advances in vaccine and therapeutic drug development have provided encouraging data concerning treatment of these infections. However, relatively little is known about immune responses in fatal versus non-fatal filovirus infection. This review summarizes the published literature on correlates of immunity to filovirus infection, and highlights deficiencies in our knowledge on this topic. It is likely that there are several types of successful immune responses, depending on the type of filovirus, and the presence and timing of vaccination or drug treatment.
Collapse
Affiliation(s)
- Steven B Bradfute
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Maryland, MD 21702, USA.
| | | |
Collapse
|
44
|
de Wit E, Feldmann H, Munster VJ. Tackling Ebola: new insights into prophylactic and therapeutic intervention strategies. Genome Med 2011; 3:5. [PMID: 21349211 PMCID: PMC3092090 DOI: 10.1186/gm219] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since its discovery in 1976, Ebolavirus has caused periodic outbreaks of viral hemorrhagic fever associated with severe and often fatal disease. Ebolavirus is endemic in Central Africa and the Philippines. Although there is currently no approved treatment available, the past 10 years has seen remarkable progress in our understanding of the pathogenicity of Ebolavirus and the development of prophylactic and post-exposure therapies against it. In vitro and in vivo experiments have shown that Ebolavirus pathogenicity is multifactorial, including viral and host determinants. Besides their function in the virus replication cycle, the viral glycoprotein, nucleoprotein, minor matrix protein and polymerase cofactor are viral determinants of pathogenicity, with evasion of the host innate and adaptive immune responses as the main mechanism. Although no licensed Ebolavirus vaccines are currently available, vaccine research in non-human primates, the 'gold standard' animal model for Ebolavirus, has produced several promising candidates. A combination of DNA vaccination and a recombinant adenovirus serotype 5 boost resulted in cross-protective immunity in non-human primates. A recombinant vesicular stomatitis vaccine vector protected non-human primates in pre- and post-exposure challenge studies. Several antiviral therapies are currently under investigation, but only a few of these have been tested in non-human primate models. Antisense therapies, in which oligonucleotides inhibit viral replication, have shown promising results in non-human primates following post-exposure treatment. In light of the severity of Ebolavirus disease and the observed increase in Ebolavirus outbreaks over the past decade, the expedited translation of potential candidate therapeutics and vaccines from bench to bedside is currently the most challenging task for the field. Here, we review the current state of Ebolavirus research, with emphasis on prophylactic and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, 903 South 4th Street, MT 59840, USA.
| | | | | |
Collapse
|
45
|
Cunningham AL, Donaghy H, Harman AN, Kim M, Turville SG. Manipulation of dendritic cell function by viruses. Curr Opin Microbiol 2010; 13:524-9. [PMID: 20598938 DOI: 10.1016/j.mib.2010.06.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 06/04/2010] [Accepted: 06/05/2010] [Indexed: 01/19/2023]
Abstract
Viruses manipulate the function of dendritic cells (DCs) to enhance their entry, spread, survival and transmission. This review summarises recently published work identifying how viruses alter the expression of receptors, antiviral molecules, disrupt signalling pathways, subvert trafficking pathways and even affect DC function via interactions with second or third cell types. Different viruses such as human immunodeficiency virus (HIV) and herpes viruses may have widely divergent and even opposite effects on DC function, determined by the need for transfer to a primary target cell, replication within the DC or various immunoevasive mechanisms.
Collapse
Affiliation(s)
- Anthony L Cunningham
- Centre for Virus Research, Westmead Millennium Institute, University of Sydney, Westmead, NSW 2145, Australia.
| | | | | | | | | |
Collapse
|
46
|
Microbial induction of vascular pathology in the CNS. J Neuroimmune Pharmacol 2010; 5:370-86. [PMID: 20401700 DOI: 10.1007/s11481-010-9208-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 03/10/2010] [Indexed: 12/31/2022]
Abstract
The central nervous system (CNS) is a finely tuned organ that participates in nearly every aspect of our day-to-day function. Neurons lie at the core of this functional unit and maintain an active dialogue with one another as well as their fellow CNS residents (e.g. astrocytes, oligodendrocytes, microglia). Because of this complex dialogue, it is essential that the CNS milieu be tightly regulated in order to permit uninterrupted and efficient neural chemistry. This is accomplished in part by anatomical barriers that segregate vascular components from the cerebral spinal fluid (CSF) and brain parenchyma. These barriers impede entry of noxious materials and enable the CNS to maintain requisite protein and ionic balances for constant electrochemical signaling. Under homeostatic conditions, the CNS is protected by the presence of specialized endothelium/epithelium, the blood brain barrier (BBB), and the blood-CSF barrier. However, following CNS infection these protective barriers can be comprised, sometimes resulting in severe neurological complications triggered by an imbalance or blockage of neural chemistry. In some instances, these disruptions are severe enough to be fatal. This review focuses on a selection of microbes (both viruses and parasites) that compromise vascular barriers and induce neurological complications upon gaining access to the CNS. Emphasis is placed on CNS diseases that result from a pathogenic interplay between host immune defenses and the invading microbe.
Collapse
|
47
|
Cárdenas WB. Evasion of the interferon-mediated antiviral response by filoviruses. Viruses 2010; 2:262-282. [PMID: 21994610 PMCID: PMC3185555 DOI: 10.3390/v2010262] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 01/11/2010] [Accepted: 01/19/2010] [Indexed: 01/09/2023] Open
Abstract
The members of the filoviruses are recognized as some of the most lethal viruses affecting human and non-human primates. The only two genera of the Filoviridae family, Marburg virus (MARV) and Ebola virus (EBOV), comprise the main etiologic agents of severe hemorrhagic fever outbreaks in central Africa, with case fatality rates ranging from 25 to 90%. Fatal outcomes have been associated with a late and dysregulated immune response to infection, very likely due to the virus targeting key host immune cells, such as macrophages and dendritic cells (DCs) that are necessary to mediate effective innate and adaptive immune responses. Despite major progress in the development of vaccine candidates for filovirus infections, a licensed vaccine or therapy for human use is still not available. During the last ten years, important progress has been made in understanding the molecular mechanisms of filovirus pathogenesis. Several lines of evidence implicate the impairment of the host interferon (IFN) antiviral innate immune response by MARV or EBOV as an important determinant of virulence. In vitro and in vivo experimental infections with recombinant Zaire Ebola virus (ZEBOV), the best characterized filovirus, demonstrated that the viral protein VP35 plays a key role in inhibiting the production of IFN-α/β. Further, the action of VP35 is synergized by the inhibition of cellular responses to IFN-α/β by the minor matrix viral protein VP24. The dual action of these viral proteins may contribute to an efficient initial virus replication and dissemination in the host. Noticeably, the analogous function of these viral proteins in MARV has not been reported. Because the IFN response is a major component of the innate immune response to virus infection, this chapter reviews recent findings on the molecular mechanisms of IFN-mediated antiviral evasion by filovirus infection.
Collapse
Affiliation(s)
- Washington B Cárdenas
- Laboratorio de Biomedicina, FIMCM, Escuela Superior Politécnica del Litoral (ESPOL), Campus Gustavo Galindo, Km 30.5 via Perimetral, Apartado 09-01-5863, Guayaquil, Ecuador
| |
Collapse
|