101
|
Abstract
Ebola virus (EBOV) infects humans as well as several animal species. It can lead to a highly lethal disease, with mortality rates approaching 90% in primates. Recent advances have deepened our understanding of how this virus is able to prevent the development of protective immune responses. The EBOV genome encodes eight proteins, four of which were shown to interact with the host in ways that counteract the immune response. The viral protein 35 (VP35) is capable of capping dsRNA and interacts with IRF7 to prevent detection of the virus by immune cells. The main role of the soluble glycoprotein (sGP) is still unclear, but it is capable of subverting the anti-GP1,2 antibody response. The GP1,2 protein has shown anti-tetherin activity and the ability to hide cell-surface proteins. Finally, VP24 interferes with the production of interferons (IFNs) and with IFN signaling in infected cells. Taken together, these data point to extensive adaptation of EBOV to evade the immune system of dead end hosts. While our understanding of the interactions between the human and viral proteins increases, details of those interactions in other hosts remain largely unclear and represent a gap in our knowledge.
Collapse
Affiliation(s)
- Jonathan Audet
- 1 Department of Medical Microbiology, University of Manitoba , Winnipeg, Manitoba, Canada
| | | |
Collapse
|
102
|
Sharma N, Cappell MS. Gastrointestinal and Hepatic Manifestations of Ebola Virus Infection. Dig Dis Sci 2015; 60:2590-2603. [PMID: 25972150 DOI: 10.1007/s10620-015-3691-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/28/2015] [Indexed: 12/09/2022]
Abstract
AIM Although Ebola virus infection (EVI) clinically presents with common, prominent, gastroenterologic manifestations, this subject has not been previously reviewed. This work critically and comprehensively reviews this subject. METHODS This study is a comprehensive literature review generated by computerized search of literature, supplemented by review of monographs and textbooks in pathology, gastroenterology, infectious diseases, and virology. RESULTS Common gastrointestinal manifestations include diarrhea-70 %, nausea and vomiting-60 %, and abdominal pain-45 %. The diarrhea and nausea and vomiting frequently produce profound, life-threatening hypovolemia requiring intravenous administration of crystalloid solutions, and frequently produce electrolyte disorders requiring electrolyte supplementation. Although gastrointestinal hemorrhage was commonly reported in early epidemics, its frequency has decreased to 10 % with prevention of disseminated intravascular coagulation. Hyperamylasemia is commonly reported, but the frequency of pancreatitis is unknown. The mean serum AST and ALT levels are each about 200/UL, with an unusual pattern for viral hepatitis of AST > ALT. The serum alkaline phosphatase averages about 160 IU/L, whereas the total bilirubin averages about 0.8 mg/dL. Risks of contracting infection during endoscopy performed on infected patients are unknown, but may be significant, as indicated by hundreds of healthcare workers contracting EVI during epidemics before instituting strict infectious control measures and anecdotal evidence of one endoscopist contracting EVI from performing endoscopy on an infected patient. CONCLUSIONS Physicians must be vigilant for gastroenterologic manifestations of EVI for appropriate diagnosis and therapy. This work should stimulate clinicopathologic studies to improve the current understanding of the gastroenterologic pathophysiology. Endoscopy is currently not standardly recommended to evaluate diarrhea, nausea and vomiting, or abdominal pain associated with EVI due to potential risks, but may be considered for endoscopic therapy for active, life-threatening, GI hemorrhage.
Collapse
Affiliation(s)
- Nisha Sharma
- Department of Medicine, Westchester Medical Center, Valhalla, NY, USA
| | | |
Collapse
|
103
|
Suppressor of Cytokine Signaling 3 Is an Inducible Host Factor That Regulates Virus Egress during Ebola Virus Infection. J Virol 2015; 89:10399-406. [PMID: 26246577 DOI: 10.1128/jvi.01736-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/28/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Ebola virus (EBOV) initially targets monocytes and macrophages, which can lead to the release of proinflammatory cytokines and chemokines. These inflammatory cytokines are thought to contribute to the development of circulatory shock seen in fatal EBOV infections. The VP40 matrix protein is a key viral structural protein that is critical for virion egress. Physical and functional interactions between VP40 and host proteins such as Tsg101 and Nedd4 facilitate efficient release of VP40-driven virus-like particles (VLPs) and infectious virus. Here, we show that host suppressor of cytokine signaling 3 (SOCS3) can also bind to EBOV VP40, leading to enhanced ubiquitinylation and egress of VP40. Indeed, titers of infectious EBOV derived from SOCS3 knockout mouse embryonic fibroblasts (MEFs) were significantly reduced compared to those from wild-type (WT) MEFs at 24 and 48 h postinfection. Importantly, this reduced virus yield could be rescued back to WT levels by exogenously expressing SOCS3. Lastly, we show that SOCS3 expression is induced by EBOV glycoprotein (GP) expression and that VLPs containing EBOV VP40 and GP induced production of proinflammatory cytokines, which induced SOCS3 for negative-feedback regulation. These data indicate that host innate immune protein SOCS3 may play an important role in budding and pathogenesis of EBOV. IMPORTANCE The VP40 matrix protein is a key structural protein critical for Ebola virus budding. Physical and functional interactions between VP40 and host proteins such as Tsg101 and Nedd4 facilitate efficient release of VLPs and infectious virus. We reported that host TLR4 is a sensor for Ebola GP on VLPs and that the resultant TLR4 signaling pathways lead to the production of proinflammatory cytokines. Host SOCS3 regulates the innate immune response by controlling and limiting the proinflammatory response through negative-feedback inhibition of cytokine receptors. We present evidence that Ebola virus VLPs stimulate induction of SOCS3 as well as proinflammatory cytokines, and that expression of human SOCS3 enhances budding of Ebola VLPs and infectious virus via a mechanism linked to the host ubiquitinylation machinery.
Collapse
|
104
|
Smither SJ, Nelson M, Eastaugh L, Nunez A, Salguero FJ, Lever MS. Experimental Respiratory Infection of Marmosets (Callithrix jacchus) With Ebola Virus Kikwit. J Infect Dis 2015. [PMID: 26209682 DOI: 10.1093/infdis/jiv371] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ebola virus (EBOV) causes a highly infectious and lethal hemorrhagic fever in primates with high fatality rates during outbreaks and EBOV may be exploited as a potential biothreat pathogen. There is therefore a need to develop and license appropriate medical countermeasures against this virus. To determine whether the common marmoset (Callithrix jacchus) would be an appropriate model to assess vaccines or therapies against EBOV disease (EVD), initial susceptibility, lethality and pathogenesis studies were performed. Low doses of EBOV-Kikwit, between 4 and 27 times the 50% tissue culture infectious dose, were sufficient to cause a lethal, reproducible infection. Animals became febrile between days 5 and 6, maintaining a high fever before succumbing to EVD between 6 and 8 days after challenge. Typical signs of EVD were observed. Pathogenesis studies revealed that virus was isolated from the lungs of animals beginning on day 3 after challenge and from the liver, spleen and blood beginning on day 5. The most striking features were observed in animals that succumbed to infection, including high viral titers in all organs, increased levels of liver function enzymes and blood clotting times, decreased levels of platelets, multifocal moderate to severe hepatitis, and perivascular edema.
Collapse
Affiliation(s)
- Sophie J Smither
- Microbiology Group, CBR Division, Defence Science and Technology Laboratory (Dstl), Salisbury
| | - Michelle Nelson
- Microbiology Group, CBR Division, Defence Science and Technology Laboratory (Dstl), Salisbury
| | - Lin Eastaugh
- Microbiology Group, CBR Division, Defence Science and Technology Laboratory (Dstl), Salisbury
| | - Alejandro Nunez
- Pathology Department, Veterinary Laboratories Agency-Weybridge, Addlestone
| | - Francisco J Salguero
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Mark S Lever
- Microbiology Group, CBR Division, Defence Science and Technology Laboratory (Dstl), Salisbury
| |
Collapse
|
105
|
Papa A, Tsergouli K, Çağlayık DY, Bino S, Como N, Uyar Y, Korukluoglu G. Cytokines as biomarkers of Crimean-Congo hemorrhagic fever. J Med Virol 2015; 88:21-7. [PMID: 26118413 DOI: 10.1002/jmv.24312] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2015] [Indexed: 01/23/2023]
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a potentially severe disease caused by CCHF virus. As in other viral hemorrhagic fevers, it is considered that the course and outcome of the disease depend on the viral load and the balance among the immune response mediators, and that a fatal outcome is the result of a "cytokine storm." The level of 27 cytokines was measured in serum samples taken from 29 patients during the acute phase of the disease. Two cases were fatal. Among survivors, significant differences between severe and non-severe cases were observed in the levels of IP-10, and MCP-1, while the levels of IL-1b, IL-5, IL-6, IL-8, IL-9, IL-10, IL-15, IP-10, MCP-1, TNF-α, and RANTES differed significantly between fatal and non-fatal cases (P < 0.05). RANTES was negatively correlated with the outcome of the disease. A striking similarity with the cytokine patterns seen in Ebola virus disease was observed. A weak Th1 immune response was seen. The viral load was positively correlated with IL-10, IP-10, and MCP-1 levels, and negatively correlated with the ratio IL-12/IL-10. Especially IP-10 and MCP-1 were significantly associated with the viral load, the severity and outcome of the disease, and they could act as biomarkers and, probably, as potential targets for treatment strategies design.
Collapse
Affiliation(s)
- Anna Papa
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Tsergouli
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dilek Yağcı Çağlayık
- Department of Microbiology Reference Laboratories, Virology Reference and Research Laboratory, Public Health Institutions of Turkey, Ankara, Turkey
| | | | - Najada Como
- Clinic of Infectious Diseases, Mother Theresa University Hospital, Tirana, Albania
| | - Yavuz Uyar
- Department of Microbiology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Gulay Korukluoglu
- Department of Microbiology Reference Laboratories, Virology Reference and Research Laboratory, Public Health Institutions of Turkey, Ankara, Turkey
| |
Collapse
|
106
|
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
|
107
|
Schirrmacher V. Signaling through RIG-I and type I interferon receptor: Immune activation by Newcastle disease virus in man versus immune evasion by Ebola virus (Review). Int J Mol Med 2015; 36:3-10. [PMID: 25998621 DOI: 10.3892/ijmm.2015.2213] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/24/2015] [Indexed: 11/06/2022] Open
Abstract
In this review, two types of RNA viruses are compared with regard to the type I interferon (IFN) response in order to obtain a better understanding of the molecular mechanisms of immune activation or evasion. Upon human infection, both viruses exert either beneficial or detrimental effects. The Newcastle disease virus (NDV), is a type strain for avian paramyxoviruses, while the Ebola virus (EBOV), is a virus affecting primates. During evolution, both viruses specifically adapted to their respective hosts, acquiring sophisticated viral escape mechanisms. Two types of receptors play an important role in the life cycle of these two viruses: cytoplasmic retinoic acid‑inducible gene I (RIG‑I) and membrane expressed type I IFN receptor (IFNAR). In mouse and human cells, NDV is a strong inducer of the type I IFN response. The early phase of this is initiated by signaling through RIG‑I and the late response by signaling through IFNAR. EBOV does not induce type I IFN responses in humans as it has viral proteins that specifically and strongly interfere with RIG‑I and IFNAR signaling, as well as immune activation. In this review, we discuss whether the beneficial effects of one virus can be exploited in the fight against the detrimental effects of the other.
Collapse
Affiliation(s)
- Volker Schirrmacher
- Division of Tumorimmunology, The Immunological and Oncological Center of Cologne, D‑50674 Cologne, Germany
| |
Collapse
|
108
|
Roberts KK, Hill TE, Davis MN, Holbrook MR, Freiberg AN. Cytokine response in mouse bone marrow derived macrophages after infection with pathogenic and non-pathogenic Rift Valley fever virus. J Gen Virol 2015; 96:1651-1663. [PMID: 25759029 PMCID: PMC4635452 DOI: 10.1099/vir.0.000119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/09/2015] [Indexed: 12/23/2022] Open
Abstract
Rift Valley fever virus (RVFV) is the most pathogenic member of the genus Phlebovirus within the family Bunyaviridae, and can cause severe disease in humans and livestock. Until recently, limited information has been published on the cellular host response elicited by RVFV, particularly in macrophages and dendritic cells, which play critical roles in stimulating adaptive and innate immune responses to viral infection. In an effort to define the initial response of host immunomodulatory cells to infection, primary mouse bone marrow derived macrophages (BMDM) were infected with the pathogenic RVFV strain ZH501, or attenuated strains MP-12 or MP-12 based Clone13 type (rMP12-C13 type), and cytokine secretion profiles examined. The secretion of T helper (Th)1-associated antiviral cytokines, chemokines and various interleukins increased rapidly after infection with the attenuated rMP12-C13 type RVFV, which lacks a functional NSs virulence gene. In comparison, infection with live-attenuated MP-12 encoding a functional NSs gene appeared to cause a delayed immune response, while pathogenic ZH501 ablates the immune response almost entirely. These data demonstrate that NSs can inhibit components of the BMDM antiviral response and supports previous work indicating that NSs can specifically regulate the type I interferon response in macrophages. Furthermore, our data demonstrate that genetic differences between ZH501 and MP-12 reduce the ability of MP-12 to inhibit antiviral signalling and subsequently reduce virulence in BMDM, demonstrating that viral components other than NSs play a critical role in regulating the host response to RVFV infection.
Collapse
Affiliation(s)
- Kimberly K. Roberts
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Terence E. Hill
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Melissa N. Davis
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Michael R. Holbrook
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Integrated Research Facility, National Institute of Allergy and Infectious Disease, National Institutes of Health, Frederick, MD, USA
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
109
|
Madariaga MG. Ebola Virus Disease: A Perspective for the United States. Am J Med 2015; 128:682-91. [PMID: 25731139 DOI: 10.1016/j.amjmed.2015.01.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 11/10/2014] [Accepted: 01/22/2015] [Indexed: 11/17/2022]
Abstract
Ebola virus caused an epidemic of unprecedented extension in West Africa. There was concern that the outbreak would not be controlled for a prolonged period of time. Two cases of infected returning travelers have been reported in the US. One of the cases has been associated with secondary transmission and other infected subjects have been repatriated for treatment. This article reviews the etiology, pathogenesis, transmission, clinical manifestations, diagnosis, treatment, and prevention of the disease with emphasis on the identification and management in the US.
Collapse
|
110
|
Abstract
Viral haemorrhagic fever can be caused by one of a diverse group of viruses that come from four different families of RNA viruses. Disease severity can vary from mild self-limiting febrile illness to severe disease characterized by high fever, high-level viraemia, increased vascular permeability that can progress to shock, multi-organ failure and death. Despite the urgent need, effective treatments and preventative vaccines are currently lacking for the majority of these viruses. A number of factors preclude the effective study of these diseases in humans including the high virulence of the agents involved, the sporadic nature of outbreaks of these viruses, which are typically in geographically isolated areas with underserviced diagnostic capabilities, and the requirements for high level bio-containment. As a result, animal models that accurately mimic human disease are essential for advancing our understanding of the pathogenesis of viral haemorrhagic fevers. Moreover, animal models for viral haemorrhagic fevers are necessary to test vaccines and therapeutic intervention strategies. Here, we present an overview of the animal models that have been established for each of the haemorrhagic fever viruses and identify which aspects of human disease are modelled. Furthermore, we discuss how experimental design considerations, such as choice of species and virus strain as well as route and dose of inoculation, have an influence on animal model development. We also bring attention to some of the pitfalls that need to be avoided when extrapolating results from animal models.
Collapse
Affiliation(s)
- D Falzaran
- Special Pathogens Programme, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - D A Bente
- Special Pathogens Programme, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.
| |
Collapse
|
111
|
Filewod NC, Lee WL. Is strengthening the endothelial barrier a therapeutic strategy for Ebola? Int J Infect Dis 2015; 36:78-9. [PMID: 26025870 DOI: 10.1016/j.ijid.2015.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 12/24/2022] Open
Affiliation(s)
- Niall C Filewod
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Warren L Lee
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
112
|
Shurtleff AC, Bavari S. Animal models for ebolavirus countermeasures discovery: what defines a useful model? Expert Opin Drug Discov 2015; 10:685-702. [PMID: 26004783 DOI: 10.1517/17460441.2015.1035252] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Ebolaviruses are highly pathogenic filoviruses, which cause disease in humans and nonhuman primates (NHP) in Africa. The Zaire ebolavirus outbreak in 2014, which continues to greatly affect Western Africa and other countries to which the hemorrhagic fever was exported due to travel of unsymptomatic yet infected individuals, was complicated by the lack of available licensed vaccines or therapeutics to combat infection. After almost a year of research at an increased pace to find and test vaccines and therapeutics, there is now a deeper understanding of the available disease models for ebolavirus infection. Demonstration of vaccine or therapeutic efficacy in NHP models of ebolavirus infection is crucial to the development and eventual licensure of ebolavirus medical countermeasures, so that safe and effective countermeasures can be accelerated into human clinical trials. AREAS COVERED The authors describe ebolavirus hemorrhagic fever (EHF) disease in various animal species: mice, guinea pigs, hamsters, pigs and NHP, to include baboons, marmosets, rhesus and cynomolgus macaques, as well as African green monkeys. Because the NHP models are supremely useful for therapeutics and vaccine testing, emphasis is placed on comparison of these models, and their use as gold-standard models of EHF. EXPERT OPINION Animal models of EHF varying from rodents to NHP species are currently under evaluation for their reproducibility and utility for modeling infection in humans. Complete development and licensure of therapeutic agents and vaccines will require demonstration that mechanisms conferring protection in NHP models of infection are predictive of protective responses in humans, for a given countermeasure.
Collapse
Affiliation(s)
- Amy C Shurtleff
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Division of Molecular and Translational Sciences , 1425 Porter Street, Frederick, MD 21702 , USA +1 301 619 4246 ; +1 541 754 3545 ;
| | | |
Collapse
|
113
|
Twenty-one days of isolation: A prospective observational cohort study of an Ebola-exposed hot zone community in Liberia. J Infect 2015; 71:150-7. [PMID: 25982026 DOI: 10.1016/j.jinf.2015.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/07/2015] [Accepted: 05/09/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND As West Africa continues to suffer from a deadly Ebola epidemic, the national health sectors struggle to minimize the damages and stop the spread of disease. METHODS A cohort of inhabitants of a small village and an Ebola hot zone in Sinoe County of Liberia was followed on a day-by-day basis to search for new cases and to minimize the spread of Ebola to the other community members or to other regions. Technical, clinical, and humanistic aspects of the response are discussed in this report. RESULTS Of the 22 confirmed Ebola cases in Sinoe County since the beginning of outbreak (June 16, 2014), 7 cases were inhabitants of Polay Town, a small village 5.5 miles east of Greenville, the Sinoe County capital. After the last wave of outbreak at the beginning of December, enhanced response activity provided essential coordination and mobilized the resources to stop the epidemic. Despite unprotected contacts in crowded houses, no new cases were detected among the contact families, or in the surrounding houses or communities. CONCLUSIONS Strong national mobilization in a decentralized but harmonized system at the community level has been of great value in controlling the epidemic in Liberia. The major interventions include epidemiological surveillance, public information dissemination, effective communication, case management, and infection control.
Collapse
|
114
|
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: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/17/2015] [Indexed: 01/07/2023]
|
115
|
Zhao JM, Dong SJ, Li J, Ji JS. The Ebola epidemic is ongoing in West Africa and responses from China are positive. Mil Med Res 2015; 2:9. [PMID: 25914830 PMCID: PMC4409763 DOI: 10.1186/s40779-015-0031-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/08/2015] [Indexed: 11/10/2022] Open
Abstract
The ongoing Ebola outbreak poses an alarming risk to the countries of West Africa and beyond. On August 8, 2014, the World Health Organization (WHO) declared the cross-country Ebola outbreak a Public Emergency of International Concern. China has had no confirmed cases of Ebola. In this paper, virologic characteristics, pathogenesis, clinical manifestations, laboratory examination and prophylactic vaccines and therapeutic drugs of Ebola are summarized. Importantly, active responses and actions from China are introduced. Moreover, the key issues in the future prevention and control of Ebola were also addressed.
Collapse
Affiliation(s)
- Jing-Min Zhao
- />Department of Pathology and Hepatology, Beijing 302 Hospital, Beijing, 100039 China
| | - Shi-Jun Dong
- />Center of Medical Information, Beijing 302 Hospital, Beijing, 100039 China
| | - Jin Li
- />Department of Infectious Diseases, Beijing 302 Hospital, Beijing, 100039 China
| | - Jun-Sheng Ji
- />Department of Infectious Diseases, Beijing 302 Hospital, Beijing, 100039 China
| |
Collapse
|
116
|
Martines RB, Ng DL, Greer PW, Rollin PE, Zaki SR. Tissue and cellular tropism, pathology and pathogenesis of Ebola and Marburg viruses. J Pathol 2015; 235:153-74. [PMID: 25297522 DOI: 10.1002/path.4456] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 09/30/2014] [Accepted: 10/01/2014] [Indexed: 12/19/2022]
Abstract
Ebola viruses and Marburg viruses include some of the most virulent and fatal pathogens known to humans. These viruses cause severe haemorrhagic fevers, with case fatality rates in the range 25-90%. The diagnosis of filovirus using formalin-fixed tissues from fatal cases poses a significant challenge. The most characteristic histopathological findings are seen in the liver; however, the findings overlap with many other viral and non-viral haemorrhagic diseases. The need to distinguish filovirus infections from other haemorrhagic fevers, particularly in areas with multiple endemic viral haemorrhagic agents, is of paramount importance. In this review we discuss the current state of knowledge of filovirus infections and their pathogenesis, including histopathological findings, epidemiology, modes of transmission and filovirus entry and spread within host organisms. The pathogenesis of filovirus infections is complex and involves activation of the mononuclear phagocytic system, with release of pro-inflammatory cytokines, chemokines and growth factors, endothelial dysfunction, alterations of the innate and adaptive immune systems, direct organ and endothelial damage from unrestricted viral replication late in infection, and coagulopathy. Although our understanding of the pathogenesis of filovirus infections has rapidly increased in the past few years, many questions remain unanswered.
Collapse
Affiliation(s)
- Roosecelis Brasil Martines
- Infectious Diseases Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
117
|
Chiappelli F, Bakhordarian A, Thames AD, Du AM, Jan AL, Nahcivan M, Nguyen MT, Sama N, Manfrini E, Piva F, Rocha RM, Maida CA. Ebola: translational science considerations. J Transl Med 2015; 13:11. [PMID: 25592846 PMCID: PMC4320629 DOI: 10.1186/s12967-014-0362-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 12/11/2014] [Indexed: 12/13/2022] Open
Abstract
We are currently in the midst of the most aggressive and fulminating outbreak of Ebola-related disease, commonly referred to as "Ebola", ever recorded. In less than a year, the Ebola virus (EBOV, Zaire ebolavirus species) has infected over 10,000 people, indiscriminately of gender or age, with a fatality rate of about 50%. Whereas at its onset this Ebola outbreak was limited to three countries in West Africa (Guinea, where it was first reported in late March 2014, Liberia, where it has been most rampant in its capital city, Monrovia and other metropolitan cities, and Sierra Leone), cases were later reported in Nigeria, Mali and Senegal, as well as in Western Europe (i.e., Madrid, Spain) and the US (i.e., Dallas, Texas; New York City) by late October 2014. World and US health agencies declared that the current Ebola virus disease (EVD) outbreak has a strong likelihood of growing exponentially across the world before an effective vaccine, treatment or cure can be developed, tested, validated and distributed widely. In the meantime, the spread of the disease may rapidly evolve from an epidemics to a full-blown pandemic. The scientific and healthcare communities actively research and define an emerging kaleidoscope of knowledge about critical translational research parameters, including the virology of EBOV, the molecular biomarkers of the pathological manifestations of EVD, putative central nervous system involvement in EVD, and the cellular immune surveillance to EBOV, patient-centered anthropological and societal parameters of EVD, as well as translational effectiveness about novel putative patient-targeted vaccine and pharmaceutical interventions, which hold strong promise, if not hope, to curb this and future Ebola outbreaks. This work reviews and discusses the principal known facts about EBOV and EVD, and certain among the most interesting ongoing or future avenues of research in the field, including vaccination programs for the wild animal vectors of the virus and the disease from global translational science perspective.
Collapse
Affiliation(s)
- Francesco Chiappelli
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
- Evidence-Based Decision Practice-Based Research Network, Los Angeles, USA.
- UCLA Center for the Health Sciences 63-090, 10833 Le Conte Avenue, Los Angeles, CA, 90095-1668, USA.
| | - Andre Bakhordarian
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
- Evidence-Based Decision Practice-Based Research Network, Los Angeles, USA.
| | - April D Thames
- UCLA David Geffen School of Medicine (Psychiatry), Los Angeles, USA.
| | - Angela M Du
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
| | - Allison L Jan
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
| | - Melissa Nahcivan
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
| | - Mia T Nguyen
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
| | - Nateli Sama
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
| | | | - Francesco Piva
- Polytechnic University of the Marche Region (Odontostomatological Sciences), Ancona, Italy.
| | | | - Carl A Maida
- UCLA School of Dentistry (Oral Biology & Medicine), Los Angeles, USA.
- UCLA School of Dentistry (Public Health Dentistry), UCLA Institute of the Environment and Sustainability, UCLA Center for Tropical Research, Los Angeles, USA.
| |
Collapse
|
118
|
Abstract
![]()
The
Ebolaviruses are members of the family Filoviridae (“filoviruses”) and cause severe hemhorragic fever
with human case fatality rates as high as 90%. Infection requires
attachment of the viral particle to cells and triggering of membrane
fusion between the host and viral membranes, a process that occurs
in the host endosome and is facilitated by the envelope glycoprotein
(GP). One potential strategy for therapeutic intervention is the development
of agents (antibodies, peptides, and small molecules) that can interfere
with viral entry aspects such as attachment, uptake, priming, or membrane
fusion. This paper highlights recent developments in the discovery
and evaluation of therapeutic entry inhibitors and identifies opportunities
moving forward.
Collapse
Affiliation(s)
- Elisabeth K. Nyakatura
- Department
of Biochemistry, Albert Einstein College of Medicine, 1300 Morris
Park Avenue, Bronx, New York 10461, United States
| | - Julia C. Frei
- Department
of Biochemistry, Albert Einstein College of Medicine, 1300 Morris
Park Avenue, Bronx, New York 10461, United States
| | - Jonathan R. Lai
- Department
of Biochemistry, Albert Einstein College of Medicine, 1300 Morris
Park Avenue, Bronx, New York 10461, United States
| |
Collapse
|
119
|
Rinkenauer AC, Schubert S, Traeger A, Schubert US. The influence of polymer architecture on in vitro pDNA transfection. J Mater Chem B 2015; 3:7477-7493. [DOI: 10.1039/c5tb00782h] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In the field of polymer-based gene delivery, the tuning potential of polymers by using different architectures like graft- and star-shaped polymers as well as self-assembled block copolymers is immense. In the last years numerous new polymer designs showed enhanced transfections properties in combination with a good biocompatibility.
Collapse
Affiliation(s)
- Alexandra C. Rinkenauer
- Laboratory of Organic and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| | - Stephanie Schubert
- Jena Center for Soft Matter (JCSM)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Institute of Pharmacy
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| |
Collapse
|
120
|
Meyers L, Frawley T, Goss S, Kang C. Ebola Virus Outbreak 2014: Clinical Review for Emergency Physicians. Ann Emerg Med 2015; 65:101-8. [DOI: 10.1016/j.annemergmed.2014.10.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/02/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
|
121
|
Ramakrishnan L, Pillai MR, Nair RR. Dengue vaccine development: strategies and challenges. Viral Immunol 2014; 28:76-84. [PMID: 25494228 DOI: 10.1089/vim.2014.0093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Infection with dengue virus may result in dengue fever or a more severe outcome, such as dengue hemorrhagic syndrome/shock. Dengue virus infection poses a threat to endemic regions for four reasons: the presence of four serotypes, each with the ability to cause a similar disease outcome, including fatality; difficulties related to vector control; the lack of specific treatment; and the nonavailability of a suitable vaccine. Vaccine development is considered challenging due to the severity of the disease observed in individuals who have acquired dengue-specific immunity, either passively or actively. Therefore, the presence of vaccine-induced immunity against a particular serotype may prime an individual to severe disease on exposure to dengue virus. Vaccine development strategies include live attenuated vaccines, chimeric, DNA-based, subunit, and inactivated vaccines. Each of the candidates is in various stages of preclinical and clinical development. Issues pertaining to selection pressures, viral interaction, and safety still need to be evaluated in order to induce a complete protective immune response against all four serotypes. This review highlights the various strategies that have been employed in vaccine development, and identifies the obstacles to producing a safe and effective vaccine.
Collapse
Affiliation(s)
- Lakshmy Ramakrishnan
- 1 Laboratory Medicine and Molecular Diagnostics, Rajiv Gandhi Centre for Biotechnology , Trivandrum, India
| | | | | |
Collapse
|
122
|
Brinton MA, Di H, Vatter HA. Simian hemorrhagic fever virus: Recent advances. Virus Res 2014; 202:112-9. [PMID: 25455336 PMCID: PMC4449332 DOI: 10.1016/j.virusres.2014.11.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/19/2014] [Accepted: 11/21/2014] [Indexed: 11/28/2022]
Abstract
SHFV induces hemorrhagic fever in macaques but not in African nonhuman primates. SHFV infection of macaque but not baboon cells induces proinflammatory cytokines. Unique N- and C-terminal genes encoded by SHFV were functionally analyzed. PLP1γ can cleave at upstream sites as well as at the expected downstream site. Eight minor structural proteins are required for infectious virus production.
The simian hemorrhagic fever virus (SHFV) genome differs from those of other members of the family Arteriviridae in encoding three papain-like one proteases (PLP1α, PLP1β and PLP1γ) at the 5′ end and two adjacent sets of four minor structural proteins at the 3′ end. The catalytic Cys and His residues and cleavage sites for each of the SHFV PLP1s were predicted and their functionality was tested in in vitro transcription/translation reactions done with wildtype or mutant polyprotein constructs. Mass spectrometry analyses of selected autoproteolytic products confirmed cleavage site locations. The catalytic Cys of PLP1α is unusual in being adjacent to an Ala instead of a Typ. PLP1γ cleaves at both downstream and upstream sites. Intermediate precursor and alternative cleavage products were detected in the in vitro transcription/translation reactions but only the three mature nsp1 proteins were detected in SHFV-infected MA104 cell lysates with SHFV nsp1 protein-specific antibodies. The duplicated sets of SHFV minor structural proteins were predicted to be functionally redundant. A stable, full-length, infectious SHFV-LVR cDNA clone was constructed and a set of mutant infectious clones was generated each with the start codon of one of the minor structural proteins mutated. All eight of the minor structural proteins were found to be required for production of infectious extracellular virus. SHFV causes a fatal hemorrhagic fever in macaques but asymptomatic, persistent infections in natural hosts such as baboons. SHFV infections were compared in macrophages and myeloid dendritic cells from baboons and macaques. Virus yields were higher from macaque cells than from baboon cells. Macrophage cultures from the two types of animals differed dramatically in the percentage of cells infected. In contrast, similar percentages of myeloid dendritic cells were infected but virus replication was efficient in the macaque cells but inefficient in the baboon cells. SHFV infection induced the production of pro-inflammatory cytokines, including IL-1β, IL-6, IL-12/23(p40), TNF-α and MIP-1α, in macaque cells but not baboon cells.
Collapse
Affiliation(s)
| | - Han Di
- Georgia State University, Atlanta, GA, USA
| | | |
Collapse
|
123
|
Lai KY, Ng WYG, Cheng FF. Human Ebola virus infection in West Africa: a review of available therapeutic agents that target different steps of the life cycle of Ebola virus. Infect Dis Poverty 2014; 3:43. [PMID: 25699183 PMCID: PMC4334593 DOI: 10.1186/2049-9957-3-43] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022] Open
Abstract
The recent outbreak of the human Zaire ebolavirus (EBOV) epidemic is spiraling out of control in West Africa. Human EBOV hemorrhagic fever has a case fatality rate of up to 90%. The EBOV is classified as a biosafety level 4 pathogen and is considered a category A agent of bioterrorism by Centers for Disease Control and Prevention, with no approved therapies and vaccines available for its treatment apart from supportive care. Although several promising therapeutic agents and vaccines against EBOV are undergoing the Phase I human trial, the current epidemic might be outpacing the speed at which drugs and vaccines can be produced. Like all viruses, the EBOV largely relies on host cell factors and physiological processes for its entry, replication, and egress. We have reviewed currently available therapeutic agents that have been shown to be effective in suppressing the proliferation of the EBOV in cell cultures or animal studies. Most of the therapeutic agents in this review are directed against non-mutable targets of the host, which is independent of viral mutation. These medications are approved by the Food and Drug Administration (FDA) for the treatment of other diseases. They are available and stockpileable for immediate use. They may also have a complementary role to those therapeutic agents under development that are directed against the mutable targets of the EBOV.
Collapse
Affiliation(s)
- Kang Yiu Lai
- />Department of Intensive Care, Queen Elizabeth Hospital, HKSAR, B6, 30 Gascoigne Rd, Kowloon, Hong Kong SAR China
| | - Wing Yiu George Ng
- />Department of Intensive Care, Queen Elizabeth Hospital, HKSAR, B6, 30 Gascoigne Rd, Kowloon, Hong Kong SAR China
| | - Fan Fanny Cheng
- />Department of Medicine, Queen Elizabeth Hospital, HKSAR, Kowloon, Hong Kong SARChina
| |
Collapse
|
124
|
Escudero-Pérez B, Volchkova VA, Dolnik O, Lawrence P, Volchkov VE. Shed GP of Ebola virus triggers immune activation and increased vascular permeability. PLoS Pathog 2014; 10:e1004509. [PMID: 25412102 PMCID: PMC4239094 DOI: 10.1371/journal.ppat.1004509] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/09/2014] [Indexed: 11/19/2022] Open
Abstract
During Ebola virus (EBOV) infection a significant amount of surface glycoprotein GP is shed from infected cells in a soluble form due to cleavage by cellular metalloprotease TACE. Shed GP and non-structural secreted glycoprotein sGP, both expressed from the same GP gene, have been detected in the blood of human patients and experimentally infected animals. In this study we demonstrate that shed GP could play a particular role during EBOV infection. In effect it binds and activates non-infected dendritic cells and macrophages inducing the secretion of pro- and anti-inflammatory cytokines (TNFα, IL1β, IL6, IL8, IL12p40, and IL1-RA, IL10). Activation of these cells by shed GP correlates with the increase in surface expression of co-stimulatory molecules CD40, CD80, CD83 and CD86. Contrary to shed GP, secreted sGP activates neither DC nor macrophages while it could bind DCs. In this study, we show that shed GP activity is likely mediated through cellular toll-like receptor 4 (TLR4) and is dependent on GP glycosylation. Treatment of cells with anti-TLR4 antibody completely abolishes shed GP-induced activation of cells. We also demonstrate that shed GP activity is negated upon addition of mannose-binding sera lectin MBL, a molecule known to interact with sugar arrays present on the surface of different microorganisms. Furthermore, we highlight the ability of shed GP to affect endothelial cell function both directly and indirectly, demonstrating the interplay between shed GP, systemic cytokine release and increased vascular permeability. In conclusion, shed GP released from virus-infected cells could activate non-infected DCs and macrophages causing the massive release of pro- and anti-inflammatory cytokines and effect vascular permeability. These activities could be at the heart of the excessive and dysregulated inflammatory host reactions to infection and thus contribute to high virus pathogenicity. Ebola virus, a member of the Filoviridae family, causes lethal hemorrhagic fever in man and primates, displaying up to 90% mortality rates. Viral infection is typified by an excessive systemic inflammatory response resembling septic shock. It also damages endothelial cells and creates difficulty in coagulation, ultimately leading to haemorrhaging, organ failure and death. A unique feature of EBOV is that following infection high amounts of truncated surface GP, named shed GP, are released from infected cells and are detected in the blood of patients and experimentally infected animals. However the role of shed GP in virus replication and pathogenicity is not yet clearly defined. Here we show that shed GP released from virus-infected cells binds and activates non-infected DCs and macrophages causing the massive release of pro- and anti-inflammatory cytokines and also affects vascular permeability. These activities could be at the heart of the excessive and dysregulated inflammatory host reactions to infection and thus contribute to high virus pathogenicity.
Collapse
Affiliation(s)
- Beatriz Escudero-Pérez
- Molecular Basis of Viral Pathogenicity, CIRI, INSERM U1111- CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Valentina A. Volchkova
- Molecular Basis of Viral Pathogenicity, CIRI, INSERM U1111- CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Olga Dolnik
- Molecular Basis of Viral Pathogenicity, CIRI, INSERM U1111- CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Philip Lawrence
- Molecular Basis of Viral Pathogenicity, CIRI, INSERM U1111- CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Viktor E. Volchkov
- Molecular Basis of Viral Pathogenicity, CIRI, INSERM U1111- CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
- * E-mail:
| |
Collapse
|
125
|
Vatter HA, Donaldson EF, Huynh J, Rawlings S, Manoharan M, Legasse A, Planer S, Dickerson MF, Lewis AD, Colgin LMA, Axthelm MK, Pecotte JK, Baric RS, Wong SW, Brinton MA. A simian hemorrhagic fever virus isolate from persistently infected baboons efficiently induces hemorrhagic fever disease in Japanese macaques. Virology 2014; 474:186-98. [PMID: 25463617 PMCID: PMC4304765 DOI: 10.1016/j.virol.2014.10.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 09/19/2014] [Accepted: 10/13/2014] [Indexed: 11/10/2022]
Abstract
Simian hemorrhagic fever virus is an arterivirus that naturally infects species of African nonhuman primates causing acute or persistent asymptomatic infections. Although it was previously estimated that 1% of baboons are SHFV-positive, more than 10% of wild-caught and captive-bred baboons tested were SHFV positive and the infections persisted for more than 10 years with detectable virus in the blood (100–1000 genomes/ml). The sequences of two baboon SHFV isolates that were amplified by a single passage in primary macaque macrophages had a high degree of identity to each other as well as to the genome of SHFV-LVR, a laboratory strain isolated in the 1960s. Infection of Japanese macaques with 100 PFU of a baboon isolate consistently produced high level viremia, pro-inflammatory cytokines, elevated tissue factor levels and clinical signs indicating coagulation defects. The baboon virus isolate provides a reliable BSL2 model of viral hemorrhagic fever disease in macaques. More than 10% of wild-caught and captive-bred baboons tested are SHFV positive. Baboons remain persistently infected for longer than 10 years. The sequences of baboon SHFV isolates were similar to each other and to SHFV LVR. A baboon SHFV (100 PFU) consistently induced hemorrhagic fever disease in macaques. A reliable BSL2 macaque model of viral hemorrhagic fever disease was developed.
Collapse
Affiliation(s)
- Heather A Vatter
- Department of Biology, Georgia State University, Atlanta, GA 30302, United States
| | - Eric F Donaldson
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Jeremy Huynh
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Stephanie Rawlings
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, United States
| | - Minsha Manoharan
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, United States
| | - Alfred Legasse
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - Shannon Planer
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - Mary F Dickerson
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - Anne D Lewis
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - Lois M A Colgin
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, United States; Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - Jerilyn K Pecotte
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, United States
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Scott W Wong
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, United States; Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR 97006, United States
| | - Margo A Brinton
- Department of Biology, Georgia State University, Atlanta, GA 30302, United States.
| |
Collapse
|
126
|
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
|
127
|
Lewnard JA, Ndeffo Mbah ML, Alfaro-Murillo JA, Altice FL, Bawo L, Nyenswah TG, Galvani AP. Dynamics and control of Ebola virus transmission in Montserrado, Liberia: a mathematical modelling analysis. THE LANCET. INFECTIOUS DISEASES 2014; 14:1189-95. [PMID: 25455986 DOI: 10.1016/s1473-3099(14)70995-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND A substantial scale-up in public health response is needed to control the unprecedented Ebola virus disease (EVD) epidemic in west Africa. Current international commitments seek to expand intervention capacity in three areas: new EVD treatment centres, case ascertainment through contact tracing, and household protective kit allocation. We aimed to assess how these interventions could be applied individually and in combination to avert future EVD cases and deaths. METHODS We developed a transmission model of Ebola virus that we fitted to reported EVD cases and deaths in Montserrado County, Liberia. We used this model to assess the effectiveness of expanding EVD treatment centres, increasing case ascertainment, and allocating protective kits for controlling the outbreak in Montserrado. We varied the efficacy of protective kits from 10% to 50%. We compared intervention initiation on Oct 15, 2014, Oct 31, 2014, and Nov 15, 2014. The status quo intervention was defined in terms of case ascertainment and capacity of EVD treatment centres on Sept 23, 2014, and all behaviour and contact patterns relevant to transmission as they were occurring at that time. The primary outcome measure was the expected number of cases averted by Dec 15, 2014. FINDINGS We estimated the basic reproductive number for EVD in Montserrado to be 2·49 (95% CI 2·38-2·60). We expect that allocating 4800 additional beds at EVD treatment centres and increasing case ascertainment five-fold in November, 2014, can avert 77 312 (95% CI 68 400-85 870) cases of EVD relative to the status quo by Dec 15, 2014. Complementing these measures with protective kit allocation raises the expectation as high as 97 940 (90 096-105 606) EVD cases. If deployed by Oct 15, 2014, equivalent interventions would have been expected to avert 137 432 (129 736-145 874) cases of EVD. If delayed to Nov 15, 2014, we expect the interventions will at best avert 53 957 (46 963-60 490) EVD cases. INTERPRETATION The number of beds at EVD treatment centres needed to effectively control EVD in Montserrado substantially exceeds the 1700 pledged by the USA to west Africa. Accelerated case ascertainment is needed to maximise effectiveness of expanding the capacity of EVD treatment centres. Distributing protective kits can further augment prevention of EVD, but it is not an adequate stand-alone measure for controlling the outbreak. Our findings highlight the rapidly closing window of opportunity for controlling the outbreak and averting a catastrophic toll of EVD cases and deaths. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
- Joseph A Lewnard
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, CT, USA
| | - Martial L Ndeffo Mbah
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, CT, USA
| | - Jorge A Alfaro-Murillo
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, CT, USA
| | - Frederick L Altice
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Infectious Diseases Section, Yale University School of Medicine, New Haven, CT, USA
| | - Luke Bawo
- Ministry of Health and Social Welfare, Monrovia, Liberia
| | | | - Alison P Galvani
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Center for Infectious Disease Modeling and Analysis, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
128
|
Papa A, Mirazimi A, Köksal I, Estrada-Pena A, Feldmann H. Recent advances in research on Crimean-Congo hemorrhagic fever. J Clin Virol 2014; 64:137-43. [PMID: 25453328 DOI: 10.1016/j.jcv.2014.08.029] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 08/25/2014] [Indexed: 11/29/2022]
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is an expanding tick-borne hemorrhagic disease with increasing human and animal health impact. Immense knowledge was gained over the past 10 years mainly due to advances in molecular biology, but also driven by an increased global interest in CCHFV as an emerging/re-emerging zoonotic pathogen. In the present article, we discuss the advances in research with focus on CCHF ecology, epidemiology, pathogenesis, diagnostics, prophylaxis and treatment. Despite tremendous achievements, future activities have to concentrate on the development of vaccines and antivirals/therapeutics to combat CCHF. Vector studies need to continue for better public and animal health preparedness and response. We conclude with a roadmap for future research priorities.
Collapse
Affiliation(s)
- Anna Papa
- Department of Microbiology, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Ali Mirazimi
- Swedish Institute for Communicable Disease Control, SE 171 82 Sweden; National Veterinary Institute, SE-756 51 Sweden; Dept for Clinical and Experimental Medicine, Linkopings University, SE-581 83 Sweden
| | - Iftihar Köksal
- Karadeniz Technical University, Medical Faculty, Department of Infectious Diseases, Trabzon, Turkey
| | | | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, USA
| |
Collapse
|
129
|
Kalra S, Kelkar D, Galwankar SC, Papadimos TJ, Stawicki SP, Arquilla B, Hoey BA, Sharpe RP, Sabol D, Jahre JA. The emergence of ebola as a global health security threat: from 'lessons learned' to coordinated multilateral containment efforts. J Glob Infect Dis 2014; 6:164-77. [PMID: 25538455 PMCID: PMC4265832 DOI: 10.4103/0974-777x.145247] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
First reported in remote villages of Africa in the 1970s, the Ebolavirus was originally believed to be transmitted to people from wild animals. Ebolavirus (EBOV) causes a severe, frequently fatal hemorrhagic syndrome in humans. Each outbreak of the Ebolavirus over the last three decades has perpetuated fear and economic turmoil among the local and regional populations in Africa. Until now it has been considered a tragic malady confined largely to the isolated regions of the African continent, but it is no longer so. The frequency of outbreaks has increased since the 1970s. The 2014 Ebola outbreak in Western Africa has been the most severe in history and was declared a public health emergency by the World Health Organization. Given the widespread use of modern transportation and global travel, the EBOV is now a risk to the entire Global Village, with intercontinental transmission only an airplane flight away. Clinically, symptoms typically appear after an incubation period of approximately 11 days. A flu-like syndrome can progress to full hemorrhagic fever with multiorgan failure, and frequently, death. Diagnosis is confirmed by detection of viral antigens or Ribonucleic acid (RNA) in the blood or other body fluids. Although historically the mortality of this infection exceeded 80%, modern medicine and public health measures have been able to lower this figure and reduce the impact of EBOV on individuals and communities. The treatment involves early, aggressive supportive care with rehydration. Core interventions, including contact tracing, preventive initiatives, active surveillance, effective isolation and quarantine procedures, and timely response to patients, are essential for a successful outbreak control. These measures, combined with public health education, point-of-care diagnostics, promising new vaccine and pharmaceutical efforts, and coordinated efforts of the international community, give new hope to the Global effort to eliminate Ebola as a public health threat. Here we present a review of EBOV infection in an effort to further educate medical and political communities on what the Ebolavirus disease entails, and what efforts are recommended to treat, isolate, and eventually eliminate it.
Collapse
Affiliation(s)
- Sarathi Kalra
- St Luke's University Health Network, Bethlehem, Pennsylvania, USA
| | | | | | | | | | | | - Brian A. Hoey
- St Luke's University Health Network, Bethlehem, Pennsylvania, USA
| | | | - Donna Sabol
- St Luke's University Health Network, Bethlehem, Pennsylvania, USA
| | - Jeffrey A. Jahre
- St Luke's University Health Network, Bethlehem, Pennsylvania, USA
| |
Collapse
|
130
|
Sheng M, Zhong Y, Chen Y, Du J, Ju X, Zhao C, Zhang G, Zhang L, Liu K, Yang N, Xie P, Li D, Zhang MQ, Jiang C. Hsa-miR-1246, hsa-miR-320a and hsa-miR-196b-5p inhibitors can reduce the cytotoxicity of Ebola virus glycoprotein in vitro. SCIENCE CHINA-LIFE SCIENCES 2014; 57:959-72. [PMID: 25218824 DOI: 10.1007/s11427-014-4742-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 08/20/2014] [Indexed: 01/11/2023]
Abstract
Ebola virus (EBOV) causes a highly lethal hemorrhagic fever syndrome in humans and has been associated with mortality rates of up to 91% in Zaire, the most lethal strain. Though the viral envelope glycoprotein (GP) mediates widespread inflammation and cellular damage, these changes have mainly focused on alterations at the protein level, the role of microRNAs (miRNAs) in the molecular pathogenesis underlying this lethal disease is not fully understood. Here, we report that the mi-RNAs hsa-miR-1246, hsa-miR-320a and hsa-miR-196b-5p were induced in human umbilical vein endothelial cells (HUVECs) following expression of EBOV GP. Among the proteins encoded by predicted targets of these miRNAs, the adhesion-related molecules tissue factor pathway inhibitor (TFPI), dystroglycan1 (DAG1) and the caspase 8 and FADD-like apoptosis regulator (CFLAR) were significantly downregulated in EBOV GP-expressing HUVECs. Moreover, inhibition of hsa-miR-1246, hsa-miR-320a and hsa-miR-196b-5p, or overexpression of TFPI, DAG1 and CFLAR rescued the cell viability that was induced by EBOV GP. Our results provide a novel molecular basis for EBOV pathogenesis and may contribute to the development of strategies to protect against future EBOV pandemics.
Collapse
Affiliation(s)
- MiaoMiao Sheng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; Department of Biochemistry and Molecular Biology, Peking Union Medical College, Tsinghua University, Beijing, 100005, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Abstract
West Africa is in the midst of the largest Ebola outbreak ever; there have been over 1000 deaths and many new cases are reported each day. The World Health Organization (WHO) declared it an outbreak in March 2014 and on August 6, 2014 the WHO declared the outbreak a public health emergency of international concern. Based on the number of deaths and total number of cases reported to the WHO as of August 11, 2014, the current outbreak has an overall mortality rate of 55%. Outbreak control measures against Ebola virus disease are effective. Why then, has this outbreak been so challenging to control? Ebola is transmitted through bodily fluids and immediately attacks the immune system, then progressively attacks the major organs and the lining of blood vessels. Sierra Leone, Guinea and Liberia are small countries that have limited resources to respond to prolonged outbreaks, especially in rural areas. This has been made more challenging by the fact that health care workers are at risk of contracting Ebola virus disease. Treatment to date has been supportive, not curative and outbreak control strategies have been met with distrust due to fear and misinformation. However, important progress is being made. The international response to Ebola is gaining momentum, communication strategies have been developed to address the fear and mistrust, and promising treatments are under development, including a combination of three monoclonal antibodies that has been administered to two American Ebola infected health care workers. The National Microbiology Laboratory of the Public Health Agency of Canada (PHAC) has been supporting laboratory diagnostic efforts in West Africa and PHAC has been working with the provinces and territories and key stakeholders to ensure Canada is prepared for a potential Ebola importation.
Collapse
|
132
|
Cellular visualization of macrophage pyroptosis and interleukin-1β release in a viral hemorrhagic infection in zebrafish larvae. J Virol 2014; 88:12026-40. [PMID: 25100833 DOI: 10.1128/jvi.02056-14] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Hemorrhagic viral diseases are distributed worldwide with important pathogens, such as dengue virus or hantaviruses. The lack of adequate in vivo infection models has limited the research on viral pathogenesis and the current understanding of the underlying infection mechanisms. Although hemorrhages have been associated with the infection of endothelial cells, other cellular types could be the main targets for hemorrhagic viruses. Our objective was to take advantage of the use of zebrafish larvae in the study of viral hemorrhagic diseases, focusing on the interaction between viruses and host cells. Cellular processes, such as transendothelial migration of leukocytes, virus-induced pyroptosis of macrophages. and interleukin-1β (Il-1β) release, could be observed in individual cells, providing a deeper knowledge of the immune mechanisms implicated in the disease. Furthermore, the application of these techniques to other pathogens will improve the current knowledge of host-pathogen interactions and increase the potential for the discovery of new therapeutic targets. Importance: Pathogenic mechanisms of hemorrhagic viruses are diverse, and most of the research regarding interactions between viruses and host cells has been performed in cell lines that might not be major targets during natural infections. Thus, viral pathogenesis research has been limited because of the lack of adequate in vivo infection models. The understanding of the relative pathogenic roles of the viral agent and the host response to the infection is crucial. This will be facilitated by the establishment of in vivo infection models using organisms such as zebrafish, which allows the study of the diseases in the context of a complete individual. The use of this animal model with other pathogens could improve the current knowledge on host-pathogen interactions and increase the potential for the discovery of new therapeutic targets against diverse viral diseases.
Collapse
|
133
|
Garamszegi S, Yen JY, Honko AN, Geisbert JB, Rubins KH, Geisbert TW, Xia Y, Hensley LE, Connor JH. Transcriptional correlates of disease outcome in anticoagulant-treated non-human primates infected with ebolavirus. PLoS Negl Trop Dis 2014; 8:e3061. [PMID: 25079789 PMCID: PMC4117489 DOI: 10.1371/journal.pntd.0003061] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/18/2014] [Indexed: 01/26/2023] Open
Abstract
Ebola virus (EBOV) infection in humans and non-human primates (NHPs) is highly lethal, and there is limited understanding of the mechanisms associated with pathogenesis and survival. Here, we describe a transcriptomic analysis of NHPs that survived lethal EBOV infection, compared to NHPs that did not survive. It has been previously demonstrated that anticoagulant therapeutics increase the survival rate in EBOV-infected NHPs, and that the characteristic transcriptional profile of immune response changes in anticoagulant-treated NHPs. In order to identify transcriptional signatures that correlate with survival following EBOV infection, we compared the mRNA expression profile in peripheral blood mononuclear cells from EBOV-infected NHPs that received anticoagulant treatment, to those that did not receive treatment. We identified a small set of 20 genes that are highly confident predictors and can accurately distinguish between surviving and non-surviving animals. In addition, we identified a larger predictive signature of 238 genes that correlated with disease outcome and treatment; this latter signature was associated with a variety of host responses, such as the inflammatory response, T cell death, and inhibition of viral replication. Notably, among survival-associated genes were subsets of genes that are transcriptionally regulated by (1) CCAAT/enhancer-binding protein alpha, (2) tumor protein 53, and (3) megakaryoblastic leukemia 1 and myocardin-like protein 2. These pathways merit further investigation as potential transcriptional signatures of host immune response to EBOV infection.
Collapse
Affiliation(s)
- Sara Garamszegi
- Bioinformatics Program, Boston University, Boston, Massachusetts, University of America
| | - Judy Y. Yen
- Department of Microbiology, School of Medicine, Boston University, Boston, Massachusetts, University of America
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, University of America
| | - Anna N. Honko
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Joan B. Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathleen H. Rubins
- National Aeronautics and Space Administration, Houston, Texas, United States of America
| | - Thomas W. Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yu Xia
- Bioinformatics Program, Boston University, Boston, Massachusetts, University of America
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Lisa E. Hensley
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, United States of America
| | - John H. Connor
- Bioinformatics Program, Boston University, Boston, Massachusetts, University of America
- Department of Microbiology, School of Medicine, Boston University, Boston, Massachusetts, University of America
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, University of America
- * E-mail:
| |
Collapse
|
134
|
Qiu X, Wong G, Fernando L, Audet J, Bello A, Strong J, Alimonti JB, Kobinger GP. mAbs and Ad-vectored IFN-α therapy rescue Ebola-infected nonhuman primates when administered after the detection of viremia and symptoms. Sci Transl Med 2014; 5:207ra143. [PMID: 24132638 DOI: 10.1126/scitranslmed.3006605] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
ZMAb is a promising treatment against Ebola virus (EBOV) disease that has been shown to protect 50% (two of four) of nonhuman primates (NHPs) when administered 2 days post-infection (dpi). To extend the treatment window and improve protection, we combined ZMAb with adenovirus-vectored interferon-α (Ad-IFN) and evaluated efficacy in EBOV-infected NHPs. Seventy-five percent (three of four) and 100% (four of four) of cynomolgus and rhesus macaques survived, respectively, when treatment was initiated after detection of viremia at 3 dpi. Fifty percent (two of four) of the cynomolgus macaques survived when Ad-IFN was given at 1 dpi, followed by ZMAb starting at 4 dpi, after positive diagnosis. The treatment was able to suppress viremia reaching ~10(5) TCID50 (median tissue culture infectious dose) per milliliter, leading to survival and robust specific immune responses. This study describes conditions capable of saving 100% of EBOV-infected NHPs when initiated after the presence of detectable viremia along with symptoms.
Collapse
Affiliation(s)
- Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Brown CS, Lee MS, Leung DW, Wang T, Xu W, Luthra P, Anantpadma M, Shabman RS, Melito LM, MacMillan KS, Borek DM, Otwinowski Z, Ramanan P, Stubbs AJ, Peterson DS, Binning JM, Tonelli M, Olson MA, Davey RA, Ready JM, Basler CF, Amarasinghe GK. In silico derived small molecules bind the filovirus VP35 protein and inhibit its polymerase cofactor activity. J Mol Biol 2014; 426:2045-58. [PMID: 24495995 PMCID: PMC4163021 DOI: 10.1016/j.jmb.2014.01.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 01/25/2014] [Accepted: 01/28/2014] [Indexed: 01/13/2023]
Abstract
The Ebola virus (EBOV) genome only encodes a single viral polypeptide with enzymatic activity, the viral large (L) RNA-dependent RNA polymerase protein. However, currently, there is limited information about the L protein, which has hampered the development of antivirals. Therefore, antifiloviral therapeutic efforts must include additional targets such as protein-protein interfaces. Viral protein 35 (VP35) is multifunctional and plays important roles in viral pathogenesis, including viral mRNA synthesis and replication of the negative-sense RNA viral genome. Previous studies revealed that mutation of key basic residues within the VP35 interferon inhibitory domain (IID) results in significant EBOV attenuation, both in vitro and in vivo. In the current study, we use an experimental pipeline that includes structure-based in silico screening and biochemical and structural characterization, along with medicinal chemistry, to identify and characterize small molecules that target a binding pocket within VP35. NMR mapping experiments and high-resolution x-ray crystal structures show that select small molecules bind to a region of VP35 IID that is important for replication complex formation through interactions with the viral nucleoprotein (NP). We also tested select compounds for their ability to inhibit VP35 IID-NP interactions in vitro as well as VP35 function in a minigenome assay and EBOV replication. These results confirm the ability of compounds identified in this study to inhibit VP35-NP interactions in vitro and to impair viral replication in cell-based assays. These studies provide an initial framework to guide development of antifiloviral compounds against filoviral VP35 proteins.
Collapse
Affiliation(s)
- Craig S Brown
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA; Biochemistry Undergraduate Program, Iowa State University, Ames, IA 50011, USA
| | - Michael S Lee
- Simulation Sciences Branch, US Army Research Laboratory, Aberdeen, MD 21005, USA; Department of Cell Biology and Biochemistry, USAMRIID, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tianjiao Wang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Wei Xu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Priya Luthra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Manu Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Reed S Shabman
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lisa M Melito
- Department of Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Karen S MacMillan
- Department of Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Dominika M Borek
- Department of Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Department of Biophysics, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Center for Structural Genomics of Infectious Diseases (CSGID), Chicago, IL, USA
| | - Zbyszek Otwinowski
- Department of Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Department of Biophysics, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Center for Structural Genomics of Infectious Diseases (CSGID), Chicago, IL, USA
| | - Parameshwaran Ramanan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Biochemistry Graduate Program, Iowa State University, Ames, IA 50011, USA
| | - Alisha J Stubbs
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA; Biochemistry Undergraduate Program, Iowa State University, Ames, IA 50011, USA
| | - Dayna S Peterson
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA; Biochemistry Undergraduate Program, Iowa State University, Ames, IA 50011, USA
| | - Jennifer M Binning
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Biochemistry Graduate Program, Iowa State University, Ames, IA 50011, USA
| | - Marco Tonelli
- National Magnetic Resonance Facility at Madison, University of Wisconsin, Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Mark A Olson
- Department of Cell Biology and Biochemistry, USAMRIID, 1425 Porter St., Fort Detrick, MD 21702, USA
| | - Robert A Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Joseph M Ready
- Department of Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Christopher F Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
136
|
Johnson JC, Martinez O, Honko AN, Hensley LE, Olinger GG, Basler CF. Pyridinyl imidazole inhibitors of p38 MAP kinase impair viral entry and reduce cytokine induction by Zaire ebolavirus in human dendritic cells. Antiviral Res 2014; 107:102-9. [PMID: 24815087 DOI: 10.1016/j.antiviral.2014.04.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 01/03/2023]
Abstract
Antigen presenting cells (APCs), including macrophages and dendritic cells, are early and sustained targets of Ebola virus (EBOV) infection in vivo. Because EBOV activates mitogen-activated protein kinase (MAPK) signaling upon infection of APCs, we evaluated the effect of pyridinyl imidazole inhibitors of p38 MAPK on EBOV infection of human APCs and EBOV mediated cytokine production from human DCs. The p38 MAPK inhibitors reduced viral replication in PMA-differentiated macrophage-like human THP-1 cells with an IC50 of 4.73μM (SB202190), 8.26μM (p38kinhIII) and 8.21μM (SB203580) and primary human monocyte-derived dendritic cells (MDDCs) with an IC50 of 2.67μM (SB202190). Furthermore, cytokine production from EBOV-treated MDDCs was inhibited in a dose-dependent manner. A control pyridinyl imidazole compound failed to inhibit either EBOV infection or cytokine induction. Using an established EBOV virus-like particle (VLP) entry assay, we demonstrate that inhibitor pretreatment blocked VLP entry suggesting that the inhibitors blocked infection and replication at least in part by blocking EBOV entry. Taken together, our results indicate that pyridinyl imidazole p38 MAPK inhibitors may serve as leads for the development of therapeutics to treat EBOV infection.
Collapse
Affiliation(s)
- Joshua C Johnson
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Osvaldo Martinez
- Dept. of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Anna N Honko
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Lisa E Hensley
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Gene G Olinger
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702, United States
| | - Christopher F Basler
- Dept. of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
137
|
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
|
138
|
Affiliation(s)
- Mike Bray
- NIAID/NIH, Biodefense Clinical Research Branch, 6700A Rockledge Drive, Room 5128, Bethesda, MD 20892, USA.
| | | |
Collapse
|
139
|
Differential responses of disease-resistant and disease-susceptible primate macrophages and myeloid dendritic cells to simian hemorrhagic fever virus infection. J Virol 2013; 88:2095-106. [PMID: 24335289 DOI: 10.1128/jvi.02633-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Simian hemorrhagic fever virus (SHFV) causes a fatal hemorrhagic fever in macaques but an asymptomatic, persistent infection in baboons. To investigate factors contributing to this differential infection outcome, the targets of SHFV infection, macrophages (MΦs) and myeloid dendritic cells (mDCs), were differentiated from macaque and baboon peripheral blood monocytes and used to compare viral replication and cell responses. SHFV replicated in >90% of macaque MΦs but in only ∼10% of baboon MΦs. Although SHFV infected ∼50% of macaque and baboon mDCs, virus replication was efficient in macaque but not in baboon mDCs. Both types of macaque cultures produced higher virus yields than baboon cultures. A more efficient type I interferon response and the production of proinflammatory cytokines, including interleukin-1β (IL-1β), IL-6, IL-12/23(p40), tumor necrosis factor alpha (TNF-α), and macrophage inflammatory protein 1α (MIP-1α), in response to SHFV infection were observed in macaque but not baboon cultures, suggesting less efficient counteraction of these responses by viral proteins in macaque cells. Baboon cultures produced higher levels of IL-10 than macaque cultures both prior to and after SHFV infection. In baboon but not macaque cell cultures, SHFV infection upregulated IL-10R1, a subunit of the IL-10 receptor (IL-10R), and also SOCS3, a negative regulator of proinflammatory cytokine production. Incubation of macaque cultures with human IL-10 before and/or after SHFV infection decreased production of IL-6, IL-1β, and MIP-1α but not TNF-α, suggesting a role for IL-10 in suppressing SHFV-induced proinflammatory cytokine production in macaques.
Collapse
|
140
|
Ayithan N, Bradfute SB, Anthony SM, Stuthman KS, Dye JM, Bavari S, Bray M, Ozato K. Ebola virus-like particles stimulate type I interferons and proinflammatory cytokine expression through the toll-like receptor and interferon signaling pathways. J Interferon Cytokine Res 2013; 34:79-89. [PMID: 24102579 DOI: 10.1089/jir.2013.0035] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ebola viruses (EBOV) can cause severe hemorrhagic disease with high case fatality rates. Currently, no vaccines or therapeutics are approved for use in humans. Ebola virus-like particles (eVLP) comprising of virus protein (VP40), glycoprotein, and nucleoprotein protect rodents and nonhuman primates from lethal EBOV infection, representing as a candidate vaccine for EBOV infection. Previous reports have shown that eVLP stimulate the expression of proinflammatory cytokines in dendritic cells (DCs) and macrophages (MΦs) in vitro. However, the molecular mechanisms and signaling pathways through which eVLP induce innate immune responses remain obscure. In this study, we show that eVLP stimulate not only the expression of proinflammatory cytokines but also the expression of type I interferons (IFNs) and IFN-stimulated genes (ISGs) in murine bone marrow-derived DCs (BMDCs) and MΦs. Our data indicate that eVLP trigger host responses through toll-like receptor (TLR) pathway utilizing 2 distinct adaptors, MyD88 and TRIF. More interestingly, eVLP activated the IFN signaling pathway by inducing a set of potent antiviral ISGs. Last, eVLP and synthetic adjuvants, Poly I:C and CpG DNA, cooperatively increased the expression of cytokines and ISGs. Further supporting this synergy, eVLP when administered together with Poly I:C conferred mice enhanced protection against EBOV infection. These results indicate that eVLP stimulate early innate immune responses through TLR and type I IFN signaling pathways to protect the host from EBOV infection.
Collapse
Affiliation(s)
- Natarajan Ayithan
- 1 Program in Genomics of Differentiation, National Institute of Child Health and Human Development , National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | | | | | |
Collapse
|
141
|
Chen H, Zheng D, Abbott J, Liu L, Bartee MY, Long M, Davids J, Williams J, Feldmann H, Strong J, Grau KR, Tibbetts S, Macaulay C, McFadden G, Thoburn R, Lomas DA, Spinale FG, Virgin HW, Lucas A. Myxomavirus-derived serpin prolongs survival and reduces inflammation and hemorrhage in an unrelated lethal mouse viral infection. Antimicrob Agents Chemother 2013; 57:4114-27. [PMID: 23774438 PMCID: PMC3754305 DOI: 10.1128/aac.02594-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 05/30/2013] [Indexed: 01/10/2023] Open
Abstract
Lethal viral infections produce widespread inflammation with vascular leak, clotting, and bleeding (disseminated intravascular coagulation [DIC]), organ failure, and high mortality. Serine proteases in clot-forming (thrombotic) and clot-dissolving (thrombolytic) cascades are activated by an inflammatory cytokine storm and also can induce systemic inflammation with loss of normal serine protease inhibitor (serpin) regulation. Myxomavirus secretes a potent anti-inflammatory serpin, Serp-1, that inhibits clotting factor X (fX) and thrombolytic tissue- and urokinase-type plasminogen activators (tPA and uPA) with anti-inflammatory activity in multiple animal models. Purified serpin significantly improved survival in a murine gammaherpesvirus 68 (MHV68) infection in gamma interferon receptor (IFN-γR) knockout mice, a model for lethal inflammatory vasculitis. Treatment of MHV68-infected mice with neuroserpin, a mammalian serpin that inhibits only tPA and uPA, was ineffective. Serp-1 reduced virus load, lung hemorrhage, and aortic, lung, and colon inflammation in MHV68-infected mice and also reduced virus load. Neuroserpin suppressed a wide range of immune spleen cell responses after MHV68 infection, while Serp-1 selectively increased CD11c(+) splenocytes (macrophage and dendritic cells) and reduced CD11b(+) tissue macrophages. Serp-1 altered gene expression for coagulation and inflammatory responses, whereas neuroserpin did not. Serp-1 treatment was assessed in a second viral infection, mouse-adapted Zaire ebolavirus in wild-type BALB/c mice, with improved survival and reduced tissue necrosis. In summary, treatment with this unique myxomavirus-derived serpin suppresses systemic serine protease and innate immune responses caused by unrelated lethal viral infections (both RNA and DNA viruses), providing a potential new therapeutic approach for treatment of lethal viral sepsis.
Collapse
Affiliation(s)
- Hao Chen
- Divisions of Cardiology and Rheumatology, Department of Medicine
- Department of Molecular Genetics and Microbiology
| | - Donghang Zheng
- Divisions of Cardiology and Rheumatology, Department of Medicine
- Department of Molecular Genetics and Microbiology
| | - Jeff Abbott
- College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Liying Liu
- Divisions of Cardiology and Rheumatology, Department of Medicine
| | - Mee Y. Bartee
- Divisions of Cardiology and Rheumatology, Department of Medicine
- Department of Molecular Genetics and Microbiology
| | - Maureen Long
- College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Jennifer Davids
- Divisions of Cardiology and Rheumatology, Department of Medicine
- Department of Molecular Genetics and Microbiology
| | | | - Heinz Feldmann
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - James Strong
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | | | | | | - Grant McFadden
- Department of Molecular Genetics and Microbiology
- Viron Therapeutics, Inc., London, Ontario, Canada
| | - Robert Thoburn
- Divisions of Cardiology and Rheumatology, Department of Medicine
| | - David A. Lomas
- Division of Pulmonary Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Francis G. Spinale
- Department of Surgery, Medical University of South Carolina, Department of Cell Biology and Anatomy, South Carolina, USA
| | - Herbert W. Virgin
- Departments of Pathology and Immunology and Molecular Microbiology, Washington University, St Louis, Missouri, USA
| | - Alexandra Lucas
- Divisions of Cardiology and Rheumatology, Department of Medicine
- Department of Molecular Genetics and Microbiology
- Viron Therapeutics, Inc., London, Ontario, Canada
| |
Collapse
|
142
|
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
|
143
|
Abstract
Filoviruses cause severe hemorrhagic fever in humans with high case-fatality rates. The cellular factors exploited by filoviruses for their spread constitute potential targets for intervention, but are incompletely defined. The viral glycoprotein (GP) mediates filovirus entry into host cells. Recent studies revealed important insights into the host cell molecules engaged by GP for cellular entry. The binding of GP to cellular lectins was found to concentrate virions onto susceptible cells and might contribute to the early and sustained infection of macrophages and dendritic cells, important viral targets. Tyrosine kinase receptors were shown to promote macropinocytic uptake of filoviruses into a subset of susceptible cells without binding to GP, while interactions between GP and human T cell Ig mucin 1 (TIM-1) might contribute to filovirus infection of mucosal epithelial cells. Moreover, GP engagement of the cholesterol transporter Niemann-Pick C1 was demonstrated to be essential for GP-mediated fusion of the viral envelope with a host cell membrane. Finally, mutagenic and structural analyses defined GP domains which interact with these host cell factors. Here, we will review the recent progress in elucidating the molecular interactions underlying filovirus entry and discuss their implications for our understanding of the viral cell tropism.
Collapse
|
144
|
Abstract
Antigen-presenting cells (APCs) are critical targets of Ebola virus (EBOV) infection in vivo. However, the susceptibility of monocytes to infection is controversial. Studies indicate productive monocyte infection, and yet monocytes are also reported to be resistant to EBOV GP-mediated entry. In contrast, monocyte-derived macrophages and dendritic cells are permissive for both EBOV entry and replication. Here, freshly isolated monocytes are demonstrated to indeed be refractory to EBOV entry. However, EBOV binds monocytes, and delayed entry occurs during monocyte differentiation. Cultured monocytes spontaneously downregulate the expression of viral entry restriction factors such as interferon-inducible transmembrane proteins, while upregulating the expression of critical EBOV entry factors cathepsin B and NPC1. Moreover, these processes are accelerated by EBOV infection. Finally, ectopic expression of NPC1 is sufficient to rescue entry into an undifferentiated, normally nonpermissive monocytic cell line. These results define the molecular basis for infection of APCs and suggest means to limit APC infection.
Collapse
|
145
|
A mutation in the Ebola virus envelope glycoprotein restricts viral entry in a host species- and cell-type-specific manner. J Virol 2013; 87:3324-34. [PMID: 23302883 DOI: 10.1128/jvi.01598-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Zaire Ebola virus (EBOV) is a zoonotic pathogen that causes severe hemorrhagic fever in humans. A single viral glycoprotein (GP) mediates viral attachment and entry. Here, virus-like particle (VLP)-based entry assays demonstrate that a GP mutant, GP-F88A, which is defective for entry into a variety of human cell types, including antigen-presenting cells (APCs), such as macrophages and dendritic cells, can mediate viral entry into mouse CD11b(+) APCs. Like that of wild-type GP (GP-wt), GP-F88A-mediated entry occurs via a macropinocytosis-related pathway and requires endosomal cysteine proteases and an intact fusion peptide. Several additional hydrophobic residues lie in close proximity to GP-F88, including L111, I113, L122, and F225. GP mutants in which these residues are mutated to alanine displayed preferential and often impaired entry into several cell types, although not in a species-specific manner. Niemann-Pick C1 (NPC1) protein is an essential filovirus receptor that binds directly to GP. Overexpression of NPC1 was recently demonstrated to rescue GP-F88A-mediated entry. A quantitative enzyme-linked immunosorbent assay (ELISA) demonstrated that while the F88A mutation impairs GP binding to human NPC1 by 10-fold, it has little impact on GP binding to mouse NPC1. Interestingly, not all mouse macrophage cell lines permit GP-F88A entry. The IC-21 cell line was permissive, whereas RAW 264.7 cells were not. Quantitative reverse transcription (RT)-PCR assays demonstrate higher NPC1 levels in GP-F88A permissive IC-21 cells and mouse peritoneal macrophages than in RAW 264.7 cells. Cumulatively, these studies suggest an important role for NPC1 in the differential entry of GP-F88A into mouse versus human APCs.
Collapse
|
146
|
Twenhafel NA, Mattix ME, Johnson JC, Robinson CG, Pratt WD, Cashman KA, Wahl-Jensen V, Terry C, Olinger GG, Hensley LE, Honko AN. Pathology of experimental aerosol Zaire ebolavirus infection in rhesus macaques. Vet Pathol 2012; 50:514-29. [PMID: 23262834 DOI: 10.1177/0300985812469636] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There is limited knowledge of the pathogenesis of human ebolavirus infections and no reported human cases acquired by the aerosol route. There is a threat of ebolavirus as an aerosolized biological weapon, and this study evaluated the pathogenesis of aerosol infection in 18 rhesus macaques. Important and unique findings include early infection of the respiratory lymphoid tissues, early fibrin deposition in the splenic white pulp, and perivasculitis and vasculitis in superficial dermal blood vessels of haired skin with rash. Initial infection occurred in the respiratory lymphoid tissues, fibroblastic reticular cells, dendritic cells, alveolar macrophages, and blood monocytes. Virus spread to regional lymph nodes, where significant viral replication occurred. Virus secondarily infected many additional blood monocytes and spread from the respiratory tissues to multiple organs, including the liver and spleen. Viremia, increased temperature, lymphocytopenia, neutrophilia, thrombocytopenia, and increased alanine aminotransferase, aspartate aminotransferase, γ-glutamyl transpeptidase, total bilirubin, serum urea nitrogen, creatinine, and hypoalbuminemia were measurable mid to late infection. Infection progressed rapidly with whole-body destruction of lymphoid tissues, hepatic necrosis, vasculitis, hemorrhage, and extravascular fibrin accumulation. Hypothermia and thrombocytopenia were noted in late stages with the development of disseminated intravascular coagulation and shock. This study provides unprecedented insight into pathogenesis of human aerosol Zaire ebolavirus infection and suggests development of a medical countermeasure to aerosol infection will be a great challenge due to massive early infection of respiratory lymphoid tissues. Rhesus macaques may be used as a model of aerosol infection that will allow the development of lifesaving medical countermeasures under the Food and Drug Administration's animal rule.
Collapse
Affiliation(s)
- N A Twenhafel
- Pathology Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Fort Detrick, MD 21702-5011, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Smith LM, Hensley LE, Geisbert TW, Johnson J, Stossel A, Honko A, Yen JY, Geisbert J, Paragas J, Fritz E, Olinger G, Young HA, Rubins KH, Karp CL. Interferon-β therapy prolongs survival in rhesus macaque models of Ebola and Marburg hemorrhagic fever. J Infect Dis 2012; 208:310-8. [PMID: 23255566 DOI: 10.1093/infdis/jis921] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
There is a clear need for novel, effective therapeutic approaches to hemorrhagic fever due to filoviruses. Ebola virus hemorrhagic fever is associated with robust interferon (IFN)-α production, with plasma concentrations of IFN-α that greatly (60- to 100-fold) exceed those seen in other viral infections, but little IFN-β production. While all of the type I IFNs signal through the same receptor complex, both quantitative and qualitative differences in biological activity are observed after stimulation of the receptor complex with different type I IFNs. Taken together, this suggested potential for IFN-β therapy in filovirus infection. Here we show that early postexposure treatment with IFN-β significantly increased survival time of rhesus macaques infected with a lethal dose of Ebola virus, although it failed to alter mortality. Early treatment with IFN-β also significantly increased survival time after Marburg virus infection. IFN-β may have promise as an adjunctive postexposure therapy in filovirus infection.
Collapse
Affiliation(s)
- Lauren M Smith
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Taniguchi S, Sayama Y, Nagata N, Ikegami T, Miranda ME, Watanabe S, Iizuka I, Fukushi S, Mizutani T, Ishii Y, Saijo M, Akashi H, Yoshikawa Y, Kyuwa S, Morikawa S. Analysis of the humoral immune responses among cynomolgus macaque naturally infected with Reston virus during the 1996 outbreak in the Philippines. BMC Vet Res 2012; 8:189. [PMID: 23057674 PMCID: PMC3528628 DOI: 10.1186/1746-6148-8-189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 10/04/2012] [Indexed: 11/22/2022] Open
Abstract
Background Ebolaviruses induce lethal viral hemorrhagic fevers (VHFs) in humans and non-human primates, with the exceptions of Reston virus (RESTV), which is not pathogenic for humans. In human VHF cases, extensive analyses of the humoral immune responses in survivors and non-survivors have shown that the IgG responses to nucleoprotein (NP) and other viral proteins are associated with asymptomatic and survival outcomes, and that the neutralizing antibody responses targeting ebolaviruses glycoprotein (GP1,2) are the major indicator of protective immunity. On the other hand, the immune responses in non-human primates, especially naturally infected ones, have not yet been elucidated in detail, and the significance of the antibody responses against NP and GP1,2 in RESTV-infected cynomolgus macaques is still unclear. In this study, we analyzed the humoral immune responses of cynomolgus macaque by using serum specimens obtained from the RESTV epizootic in 1996 in the Philippines to expand our knowledge on the immune responses in naturally RESTV-infected non-human primates. Results The antibody responses were analyzed using IgG-ELISA, an indirect immunofluorescent antibody assay (IFA), and a pseudotyped VSV-based neutralizing (NT) assay. Antigen-capture (Ag)-ELISA was also performed to detect viral antigens in the serum specimens. We found that the anti-GP1,2 responses, but not the anti-NP responses, closely were correlated with the neutralization responses, as well as the clearance of viremia in the sera of the RESTV-infected cynomolgus macaques. Additionally, by analyzing the cytokine/chemokine concentrations of these serum specimens, we found high concentrations of proinflammatory cytokines/chemokines, such as IFNγ, IL8, IL-12, and MIP1α, in the convalescent phase sera. Conclusions These results imply that both the antibody response to GP1,2 and the proinflammatory innate responses play significant roles in the recovery from RESTV infection in cynomolgus macaques.
Collapse
Affiliation(s)
- Satoshi Taniguchi
- Special Pathogens Laboratory, Department of Virology 1, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo, 208-0011, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Shurtleff AC, Warren TK, Bavari S. Nonhuman primates as models for the discovery and development of ebolavirus therapeutics. Expert Opin Drug Discov 2012; 6:233-50. [PMID: 22647202 DOI: 10.1517/17460441.2011.554815] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Ebolaviruses are human pathogenic Category A priority pathogens for which no vaccines or therapeutics are currently licensed; however, several therapeutic agents have shown promising efficacy in nonhuman primate models of infection and are potential candidates for use in humans. Demonstration of efficacy in nonhuman primate models of ebolavirus infection will probably be central to the development and eventual licensure of ebolavirus medical countermeasures given the ethical and feasibility constraints of human efficacy assessments. AREAS COVERED The authors describe ebolavirus hemorrhagic fever (EHF), with an emphasis on comparing human and nonhuman primate pathophysiology. Published data examining human and animal clinical disease parameters, histopathological findings, and immune responses in fatal and nonfatal cases are synthesized and evaluated. Importantly, the authors also introduce and describe the FDA Animal Efficacy Rule as well as recent advances in antiviral drug development strategies for the treatment of EHF. EXPERT OPINION Well-characterized models of ebolavirus infection are currently under development and scrutiny as to their accuracy and utility for modeling fatal infection in humans. The advanced development and eventual licensure of therapeutic agents will require demonstration that mechanisms conferring protection in nonhuman primate models of infection are predictive of protective responses in humans.
Collapse
Affiliation(s)
- Amy C Shurtleff
- US Army Medical Research Institute of Infectious Diseases, Integrated Toxicology Division, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA +1 301 619 4246 ; +1 541 754 3545 ;
| | | | | |
Collapse
|
150
|
Paweska JT, Jansen van Vuren P, Masumu J, Leman PA, Grobbelaar AA, Birkhead M, Clift S, Swanepoel R, Kemp A. Virological and serological findings in Rousettus aegyptiacus experimentally inoculated with vero cells-adapted hogan strain of Marburg virus. PLoS One 2012; 7:e45479. [PMID: 23029039 PMCID: PMC3444458 DOI: 10.1371/journal.pone.0045479] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 08/20/2012] [Indexed: 12/16/2022] Open
Abstract
The Egyptian fruit bat, Rousettus aegyptiacus, is currently regarded as a potential reservoir host for Marburg virus (MARV). However, the modes of transmission, the level of viral replication, tissue tropism and viral shedding pattern remains to be described. Captive-bred R. aegyptiacus, including adult males, females and pups were exposed to MARV by different inoculation routes. Blood, tissues, feces and urine from 9 bats inoculated by combination of nasal and oral routes were all negative for the virus and ELISA IgG antibody could not be demonstrated for up to 21 days post inoculation (p.i.). In 21 bats inoculated by a combination of intraperitoneal/subcutaneous route, viremia and the presence of MARV in different tissues was detected on days 2-9 p.i., and IgG antibody on days 9-21 p.i. In 3 bats inoculated subcutaneously, viremia was detected on days 5 and 8 (termination of experiment), with virus isolation from different organs. MARV could not be detected in urine, feces or oral swabs in any of the 3 experimental groups. However, it was detected in tissues which might contribute to horizontal or vertical transmission, e.g. lung, intestines, kidney, bladder, salivary glands, and female reproductive tract. Viremia lasting at least 5 days could also facilitate MARV mechanical transmission by blood sucking arthropods and infections of susceptible vertebrate hosts by direct contact with infected blood. All bats were clinically normal and no gross pathology was identified on post mortem examination. This work confirms the susceptibility of R. aegyptiacus to infection with MARV irrespective of sex and age and contributes to establishing a bat-filovirus experimental model. Further studies are required to uncover the mode of MARV transmission, and to investigate the putative role of R. aegyptiacus as a reservoir host.
Collapse
Affiliation(s)
- Janusz T Paweska
- Center for Emerging and Zoonotic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Sandringham, South Africa.
| | | | | | | | | | | | | | | | | |
Collapse
|