106 results on '"Marburgvirus pathogenicity"'
Search Results
2. Marburg Virus Aerosol Infection in Animal Models.
- Author
-
Bushmaker T
- Subjects
- Animals, Mice, Humans, Marburgvirus pathogenicity, Aerosols, Marburg Virus Disease virology, Disease Models, Animal
- Abstract
Aerosol challenge provides the ability to investigate in-depth mucosal virus exposure, including modeling of airborne transmission from high-consequence viral infection. For filoviruses, in particular Marburg and Ebola viruses, aerosol infection is considered in connection to their potential misuse. Aerosol exposure with filoviruses has been used to study disease progression, pathogenicity, and the efficacy of medical countermeasures. The aim of this protocol is to provide a general outline for aerosol exposure of animals with filoviruses, specifically Marburg virus., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
3. A Hamster Model for the Evaluation of Marburg Virus Countermeasures.
- Author
-
Smith BJ and Marzi A
- Subjects
- Animals, Cricetinae, Mesocricetus, Humans, Marburgvirus immunology, Marburgvirus pathogenicity, Marburg Virus Disease virology, Marburg Virus Disease immunology, Disease Models, Animal
- Abstract
Wild-type filoviruses including Marburg virus (MARV) cause disease in humans, nonhuman primates, and some immunodeficient mouse strains but generally not in immunocompetent rodents and ferrets. However, disease in immunocompetent rodents can be achieved by serial passaging of the virus as demonstrated by the mouse-, hamster-, and guinea pig-adapted strains of MARV, which often cause lethal disease in the respective rodent species. These disease models present valuable first screening models for medical countermeasure evaluation against MARV, including monoclonal antibody therapies and vaccines. The MARV hamster disease model is of particular interest since the infected hamsters display almost all of the clinical signs of Marburg virus disease observed in nonhuman primates and humans, including petechial rash, hemorrhages, coagulation disorder, and dysregulated immune responses. This chapter describes a protocol using the hamster-adapted MARV in Syrian Golden hamsters for studies investigating viral pathogenesis or evaluating the efficacy of medical countermeasures., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
4. Prototype of a DNA Vaccine against Marburg Virus.
- Author
-
Volkova NV, Pyankov OV, Ivanova AV, Isaeva AA, Zybkina AV, Kazachinskaya EI, and Shcherbakov DN
- Subjects
- Animals, Antibodies, Neutralizing immunology, Antibodies, Neutralizing metabolism, Enzyme-Linked Immunosorbent Assay, Glycoproteins immunology, Glycoproteins metabolism, Immunoglobulins immunology, Immunoglobulins metabolism, Marburgvirus immunology, Viral Proteins immunology, Viral Proteins metabolism, Marburgvirus pathogenicity, Vaccines, DNA therapeutic use
- Abstract
The preparation and study of the biological properties of the pVAKS-GPVM DNA immunogen containing a gene encoding Marburgvirus glycoprotein are described. The specificity of blood serum antibodies of guinea pigs immunized with DNA immunogen was analyzed by ELISA. Inactivated viral preparation, recombinant glycoprotein (GP) obtained in the prokaryotic system and virus-like particles based on the recombinant vesicular stomatitis virus exhibiting Marburgvirus GP were used as the antigens. The neutralizing activity of antibodies of immunized animals was tested in vitro using a pseudovirus system. It was demonstrated that the developed immunogen administered to guinea pigs induced the production of specific antibodies that neutralize virus-like particles and Marburgvirus in cultured Vero cells.
- Published
- 2021
- Full Text
- View/download PDF
5. Discovery of Marburg virus neutralizing antibodies from virus-naïve human antibody repertoires using large-scale structural predictions.
- Author
-
Bozhanova NG, Sangha AK, Sevy AM, Gilchuk P, Huang K, Nargi RS, Reidy JX, Trivette A, Carnahan RH, Bukreyev A, Crowe JE Jr, and Meiler J
- Subjects
- Animals, Antibodies, Neutralizing genetics, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Complementarity Determining Regions immunology, Epitopes genetics, Epitopes immunology, Glycoproteins genetics, Glycoproteins immunology, Humans, Immunoglobulin Heavy Chains genetics, Immunoglobulin Heavy Chains immunology, Marburg Virus Disease drug therapy, Marburg Virus Disease genetics, Marburg Virus Disease virology, Marburgvirus pathogenicity, Mutation genetics, Mutation immunology, Viral Envelope Proteins, Viral Vaccines genetics, Viral Vaccines immunology, Antibodies, Viral genetics, Complementarity Determining Regions genetics, Marburg Virus Disease immunology, Marburgvirus immunology
- Abstract
Marburg virus (MARV) disease is lethal, with fatality rates up to 90%. Neutralizing antibodies (Abs) are promising drug candidates to prevent or treat the disease. Current efforts are focused in part on vaccine development to induce such MARV-neutralizing Abs. We analyzed the antibody repertoire from healthy unexposed and previously MARV-infected individuals to assess if naïve repertoires contain suitable precursor antibodies that could become neutralizing with a limited set of somatic mutations. We computationally searched the human Ab variable gene repertoire for predicted structural homologs of the neutralizing Ab MR78 that is specific to the receptor binding site (RBS) of MARV glycoprotein (GP). Eight Ab heavy-chain complementarity determining region 3 (HCDR3) loops from MARV-naïve individuals and one from a previously MARV-infected individual were selected for testing as HCDR3 loop chimeras on the MR78 Ab framework. Three of these chimerized antibodies bound to MARV GP. We then tested a full-length native Ab heavy chain encoding the same 17-residue-long HCDR3 loop that bound to the MARV GP the best among the chimeric Abs tested. Despite only 57% amino acid sequence identity, the Ab from a MARV-naïve donor recognized MARV GP and possessed neutralizing activity against the virus. Crystallization of both chimeric and full-length native heavy chain-containing Abs provided structural insights into the mechanism of binding for these types of Abs. Our work suggests that the MARV GP RBS is a promising candidate for epitope-focused vaccine design to induce neutralizing Abs against MARV., Competing Interests: Competing interest statement: J.E.C. is on the Scientific Advisory Boards of CompuVax and Meissa Vaccines, a recipient of previous unrelated research grants from Moderna and Sanofi, and founder of IDBiologics, Inc. Vanderbilt University has applied for a patent that includes the original MR78 Ab.
- Published
- 2020
- Full Text
- View/download PDF
6. Modeling the efficiency of filovirus entry into cells in vitro: Effects of SNP mutations in the receptor molecule.
- Author
-
Kim KS, Kondoh T, Asai Y, Takada A, and Iwami S
- Subjects
- Cell Line, Ebolavirus genetics, Ebolavirus pathogenicity, Humans, Marburgvirus genetics, Marburgvirus pathogenicity, Mutation, Polymorphism, Single Nucleotide, Viral Plaque Assay, Ebolavirus physiology, Marburgvirus physiology, Models, Biological, Virus Internalization
- Abstract
Interaction between filovirus glycoprotein (GP) and the Niemann-Pick C1 (NPC1) protein is essential for membrane fusion during virus entry. Some single-nucleotide polymorphism (SNPs) in two surface-exposed loops of NPC1 are known to reduce viral infectivity. However, the dependence of differences in entry efficiency on SNPs remains unclear. Using vesicular stomatitis virus pseudotyped with Ebola and Marburg virus GPs, we investigated the cell-to-cell spread of viruses in cultured cells expressing NPC1 or SNP derivatives. Eclipse and virus-producing phases were assessed by in vitro infection experiments, and we developed a mathematical model describing spatial-temporal virus spread. This mathematical model fit the plaque radius data well from day 2 to day 6. Based on the estimated parameters, we found that SNPs causing the P424A and D508N substitutions in NPC1 most effectively reduced the entry efficiency of Ebola and Marburg viruses, respectively. Our novel approach could be broadly applied to other virus plaque assays., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
7. Marburg virus pathogenesis - differences and similarities in humans and animal models.
- Author
-
Shifflett K and Marzi A
- Subjects
- Animals, Disease Models, Animal, Host-Pathogen Interactions, Marburg Virus Disease physiopathology, Marburgvirus growth & development, Marburgvirus pathogenicity
- Abstract
Marburg virus (MARV) is a highly pathogenic virus associated with severe disease and mortality rates as high as 90%. Outbreaks of MARV are sporadic, deadly, and often characterized by a lack of resources and facilities to diagnose and treat patients. There are currently no approved vaccines or treatments, and the chaotic and infrequent nature of outbreaks, among other factors, makes testing new countermeasures during outbreaks ethically and logistically challenging. Without field efficacy studies, researchers must rely on animal models of MARV infection to assess the efficacy of vaccines and treatments, with the limitations being the accuracy of the animal model in recapitulating human pathogenesis. This review will compare various animal models to the available descriptions of human pathogenesis and aims to evaluate their effectiveness in modeling important aspects of Marburg virus disease.
- Published
- 2019
- Full Text
- View/download PDF
8. The Role of Reference Materials in the Research and Development of Diagnostic Tools and Treatments for Haemorrhagic Fever Viruses.
- Author
-
Mattiuzzo G, Bentley EM, and Page M
- Subjects
- Africa, Western epidemiology, Animals, Antigens, Viral blood, Dengue Virus immunology, Dengue Virus isolation & purification, Dengue Virus pathogenicity, Disease Outbreaks prevention & control, Ebolavirus immunology, Ebolavirus isolation & purification, Ebolavirus pathogenicity, Epidemics prevention & control, Hemorrhagic Fever Virus, Crimean-Congo immunology, Hemorrhagic Fever Virus, Crimean-Congo isolation & purification, Hemorrhagic Fever Virus, Crimean-Congo pathogenicity, Hemorrhagic Fever, Crimean diagnosis, Hemorrhagic Fever, Crimean immunology, Hemorrhagic Fever, Crimean prevention & control, Humans, Lassa Fever diagnosis, Lassa Fever immunology, Lassa Fever prevention & control, Lassa virus immunology, Lassa virus isolation & purification, Lassa virus pathogenicity, Marburg Virus Disease diagnosis, Marburg Virus Disease immunology, Marburg Virus Disease prevention & control, Marburgvirus immunology, Marburgvirus isolation & purification, Marburgvirus pathogenicity, RNA Viruses immunology, RNA Viruses isolation & purification, RNA Viruses pathogenicity, RNA, Viral isolation & purification, Rift Valley Fever diagnosis, Rift Valley Fever immunology, Rift Valley Fever prevention & control, Rift Valley fever virus immunology, Rift Valley fever virus isolation & purification, Rift Valley fever virus pathogenicity, Severe Dengue diagnosis, Severe Dengue immunology, Severe Dengue prevention & control, World Health Organization, Diagnostic Techniques and Procedures, Hemorrhagic Fever, Ebola diagnosis, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola prevention & control, Information Services, RNA Virus Infections diagnosis, RNA Virus Infections immunology, RNA Virus Infections prevention & control, Vaccines standards
- Abstract
Following the Ebola outbreak in Western Africa in 2013-16, a global effort has taken place for preparedness for future outbreaks. As part of this response, the development of vaccines, treatments and diagnostic tools has been accelerated, especially towards pathogens listed as likely to cause an epidemic and for which there are no current treatments. Several of the priority pathogens identified by the World Health Organisation are haemorrhagic fever viruses. This review provides information on the role of reference materials as an enabling tool for the development and evaluation of assays, and ultimately vaccines and treatments. The types of standards available are described, along with how they can be applied for assay harmonisation through calibration as a relative potency to a common arbitrary unitage system (WHO International Unit). This assures that assay metrology is accurate and robust. We describe reference materials that have been or are being developed for haemorrhagic fever viruses and consider the issues surrounding their production, particularly that of biosafety where the viruses require specialised containment facilities. Finally, we advocate the use of reference materials at early stages, including research and development, as this helps produce reliable assays and can smooth the path to regulatory approval.
- Published
- 2019
- Full Text
- View/download PDF
9. Recent advances in marburgvirus research.
- Author
-
Olejnik J, Mühlberger E, and Hume AJ
- Subjects
- Africa, Western, Animals, Biomedical Research trends, Disease Models, Animal, Disease Outbreaks, Ebolavirus, Humans, Chiroptera, Marburgvirus pathogenicity
- Abstract
Marburgviruses are closely related to ebolaviruses and cause a devastating disease in humans. In 2012, we published a comprehensive review of the first 45 years of research on marburgviruses and the disease they cause, ranging from molecular biology to ecology. Spurred in part by the deadly Ebola virus outbreak in West Africa in 2013-2016, research on all filoviruses has intensified. Not meant as an introduction to marburgviruses, this article instead provides a synopsis of recent progress in marburgvirus research with a particular focus on molecular biology, advances in animal modeling, and the use of Egyptian fruit bats in infection experiments., Competing Interests: No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.
- Published
- 2019
- Full Text
- View/download PDF
10. Marburg haemorrhagic fever in returning travellers: an overview aimed at clinicians.
- Author
-
Bauer MP, Timen A, Vossen ACTM, and van Dissel JT
- Subjects
- Animals, Disease Outbreaks prevention & control, Early Diagnosis, Humans, Infection Control, Marburg Virus Disease pathology, Marburg Virus Disease therapy, Marburgvirus pathogenicity, Marburg Virus Disease diagnosis, Marburg Virus Disease prevention & control, Marburgvirus isolation & purification, Travel-Related Illness
- Abstract
Marburg virus haemorrhagic fever (MARV HF) is a dramatic disease that can occur in a traveller returning from an area where the virus is endemic. In this article, we provide an overview of MARV HF as an imported infection with an emphasis on clinical aspects. Although late features such as rash, signs of haemorrhagic diathesis and liver necrosis may point to the diagnosis, the initial clinical picture is non-specific. If in this early phase the patient's epidemiological exposure history is compatible with MARV HF, the patient should be isolated and managed according to viral haemorrhagic fever protocol and RT-PCR should be performed on the patient's blood as soon as possible to rule out MARV HF (or other possible viral haemorrhagic fevers). In severe cases, direct electron microscopy of blood in specialized centres (e.g. Bernhard-Nocht Institute in Hamburg, Germany) may be considered if the result of the RT-PCR is not readily available. Adequate diagnostics and empirical treatment for other acute life-threatening illnesses should not be withheld while test results are awaited, but all management and diagnostics should be weighed against the risks of nosocomial transmission., (Copyright © 2016. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
11. Complete protection of the BALB/c and C57BL/6J mice against Ebola and Marburg virus lethal challenges by pan-filovirus T-cell epigraph vaccine.
- Author
-
Rahim MN, Wee EG, He S, Audet J, Tierney K, Moyo N, Hannoun Z, Crook A, Baines A, Korber B, Qiu X, and Hanke T
- Subjects
- Animals, Antibodies, Neutralizing, Antibodies, Viral, Ebola Vaccines, Ebolavirus pathogenicity, Female, Filoviridae metabolism, Filoviridae pathogenicity, Hemorrhagic Fever, Ebola, Immunity, Cellular immunology, Male, Marburgvirus pathogenicity, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Proof of Concept Study, T-Lymphocytes metabolism, Filoviridae immunology, T-Lymphocytes immunology, Viral Vaccines pharmacology
- Abstract
There are a number of vaccine candidates under development against a small number of the most common outbreak filoviruses all employing the virus glycoprotein (GP) as the vaccine immunogen. However, antibodies induced by such GP vaccines are typically autologous and limited to the other members of the same species. In contrast, T-cell vaccines offer a possibility to design a single pan-filovirus vaccine protecting against all known and even likely existing, but as yet unencountered members of the family. Here, we used a cross-filovirus immunogen based on conserved regions of the filovirus nucleoprotein, matrix and polymerase to construct simian adenovirus- and poxvirus MVA-vectored vaccines, and in a proof-of-concept study demonstrated a protection of the BALB/c and C57BL/6J mice against high, lethal challenges with Ebola and Marburg viruses, two distant members of the family, by vaccine-elicited T cells in the absence of GP antibodies., Competing Interests: NO authors have competing interests.
- Published
- 2019
- Full Text
- View/download PDF
12. Marburgviruses: An Update.
- Author
-
Miraglia CM
- Subjects
- Animals, Humans, Marburg Virus Disease pathology, Marburg Virus Disease therapy, Marburg Virus Disease transmission, Marburgvirus genetics, Marburg Virus Disease epidemiology, Marburgvirus pathogenicity
- Abstract
Ebolaviruses have gained much attention recently due to the outbreak from 2014 through 2016. The related marburgviruses also have been responsible for large outbreaks with high case fatality rates. The purpose of this article is to provide the clinical laboratory scientist with a review of the most current developments in marburgvirus research. The PubMed database was reviewed using the keywords "Marburg virus," "Ravn virus," and "marburgviruses," with publication dates from January 1, 2015 through June 20, 2017. The search yielded 345 articles. In total, 52 articles met the inclusion criteria and were reviewed. Advances have been made in the areas of ecology and host reservoir studies, seroprevalence studies, pathology and pathogenesis studies, laboratory assay development, and treatment and vaccine development. Marburgviruses are highly lethal viruses that pose a significant threat to the human population. Although numerous advances have been made, there are still large gaps in knowledge, and it is imperative that scientists gain more information to fully understand virus/host interactions. An approved vaccine and treatment remain elusive.
- Published
- 2019
- Full Text
- View/download PDF
13. Distinct Biological Phenotypes of Marburg and Ravn Virus Infection in Macaques.
- Author
-
Nicholas VV, Rosenke R, Feldmann F, Long D, Thomas T, Scott DP, Feldmann H, and Marzi A
- Subjects
- Animals, Apoptosis, Hepatocytes pathology, Macaca fascicularis, Macaca mulatta, Macrophages pathology, Male, Marburg Virus Disease immunology, Marburg Virus Disease pathology, Phenotype, Marburg Virus Disease etiology, Marburgvirus pathogenicity
- Abstract
Filoviruses are among the most pathogenic infectious agents known to human, with high destructive potential, as evidenced by the recent Ebola virus epidemic in West Africa. As members of the filovirus family, marburgviruses have caused similar devastating outbreaks, albeit with lower case numbers. In this study we compare the pathogenesis of Ravn virus (RAVV) and Marburg virus (MARV) strains Angola, Musoke, and Ozolin in rhesus and cynomolgus macaques, the 2 nonhuman primate species most commonly used in filovirus research. Our results reveal the most pathogenic MARV strain to be Angola, followed by Musoke, whereas Ozolin is the least pathogenic. We also demonstrate that RAVV is highly pathogenic in cynomolgus macaques but less pathogenic in rhesus macaques. Our results demonstrate a preferential infection of endothelial cells by MARVs; in addition, analysis of tissue samples suggests that lymphocyte and hepatocyte apoptosis might play a role in MARV pathogenicity. This information expands our knowledge about pathogenicity and virulence of marburgviruses.
- Published
- 2018
- Full Text
- View/download PDF
14. Pathogenicity of Ebola and Marburg Viruses Is Associated With Differential Activation of the Myeloid Compartment in Humanized Triple Knockout-Bone Marrow, Liver, and Thymus Mice.
- Author
-
Lavender KJ, Williamson BN, Saturday G, Martellaro C, Griffin A, Hasenkrug KJ, Feldmann H, and Prescott J
- Subjects
- Animals, Bone Marrow immunology, Ebolavirus immunology, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola virology, Immunity immunology, Liver immunology, Liver virology, Macrophages immunology, Macrophages virology, Marburg Virus Disease immunology, Marburg Virus Disease virology, Marburgvirus immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Myeloid Cells immunology, Thymus Gland immunology, Virulence immunology, Bone Marrow virology, Ebolavirus pathogenicity, Marburgvirus pathogenicity, Myeloid Cells virology, Thymus Gland virology
- Abstract
Ebola virus (EBOV) and Marburg virus (MARV) outbreaks are highly lethal, and infection results in a hemorrhagic fever with complex etiology. These zoonotic viruses dysregulate the immune system to cause disease, in part by replicating within myeloid cells that would normally innately control viral infection and shape the adaptive immune response. We used triple knockout (TKO)-bone marrow, liver, thymus (BLT) humanized mice to recapitulate the early in vivo human immune response to filovirus infection. Disease severity in TKO-BLT mice was dissimilar between EBOV and MARV with greater severity observed during EBOV infection. Disease severity was related to increased Kupffer cell infection in the liver, higher levels of myeloid dysfunction, and skewing of macrophage subtypes in EBOV compared with MARV-infected mice. Overall, the TKO-BLT model provided a practical in vivo platform to study the human immune response to filovirus infection and generated a better understanding of how these viruses modulate specific components of the immune system.
- Published
- 2018
- Full Text
- View/download PDF
15. Virulence of Marburg Virus Angola Compared to Mt. Elgon (Musoke) in Macaques: A Pooled Survival Analysis.
- Author
-
Blair PW, Keshtkar-Jahromi M, Psoter KJ, Reisler RB, Warren TK, Johnston SC, Goff AJ, Downey LG, Bavari S, and Cardile AP
- Subjects
- Animals, Disease Models, Animal, Injections, Intramuscular, Retrospective Studies, Survival Analysis, United States, Virulence, Macaca, Marburg Virus Disease pathology, Marburgvirus pathogenicity
- Abstract
Angola variant (MARV/Ang) has replaced Mt. Elgon variant Musoke isolate (MARV/MtE-Mus) as the consensus standard variant for Marburg virus research and is regarded as causing a more aggressive phenotype of disease in animal models; however, there is a dearth of published evidence supporting the higher virulence of MARV/Ang. In this retrospective study, we used data pooled from eight separate studies in nonhuman primates experimentally exposed with either 1000 pfu intramuscular (IM) MARV/Ang or MARV/MtE-Mus between 2012 and 2017 at the United States Army Medical Research Institute of Infectious Diseases (USAMRIID). Multivariable Cox proportional hazards regression was used to evaluate the association of variant type with time to death, the development of anorexia, rash, viremia, and 10 select clinical laboratory values. A total of 47 cynomolgus monkeys were included, of which 18 were exposed to MARV/Ang in three separate studies and 29 to MARV/MtE-Mus in five studies. Following universally fatal Marburg virus exposure, compared to MARV/MtE-Mus, MARV/Ang was associated with an increased risk of death (HR = 22.10; 95% CI: 7.08, 68.93), rash (HR = 5.87; 95% CI: 2.76, 12.51) and loss of appetite (HR = 35.10; 95% CI: 7.60, 162.18). Our data demonstrate an increased virulence of MARV/Ang compared to MARV/MtE-Mus variant in the 1000 pfu IM cynomolgus macaque model.
- Published
- 2018
- Full Text
- View/download PDF
16. A Chimeric Lloviu Virus Minigenome System Reveals that the Bat-Derived Filovirus Replicates More Similarly to Ebolaviruses than Marburgviruses.
- Author
-
Manhart WA, Pacheco JR, Hume AJ, Cressey TN, Deflubé LR, and Mühlberger E
- Subjects
- Animals, Cell Line, Tumor, Ebolavirus genetics, Filoviridae genetics, Filoviridae pathogenicity, HEK293 Cells, Humans, Marburgvirus genetics, RNA-Dependent RNA Polymerase genetics, RNA-Dependent RNA Polymerase metabolism, Virus Replication genetics, Chiroptera virology, Ebolavirus pathogenicity, Genome, Viral genetics, Marburgvirus pathogenicity, Virus Replication physiology
- Abstract
Recently, traces of zoonotic viruses have been discovered in bats and other species around the world, but despite repeated attempts, full viral genomes have not been rescued. The absence of critical genetic sequences from these viruses and the difficulties to isolate infectious virus from specimens prevent research on their pathogenic potential for humans. One example of these zoonotic pathogens is Lloviu virus (LLOV), a filovirus that is closely related to Ebola virus. Here, we established LLOV minigenome systems based on sequence complementation from other filoviruses. Our results show that the LLOV replication and transcription mechanisms are, in general, more similar to ebolaviruses than to marburgviruses. We also show that a single nucleotide at the 3' genome end determines species specificity of the LLOV polymerase. The data obtained here will be instrumental for the rescue of infectious LLOV clones for pathogenesis studies., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. Catching Chances: The Movement to Be on the Ground and Research Ready before an Outbreak.
- Author
-
Brett-Major D and Lawler J
- Subjects
- Animals, Democratic Republic of the Congo epidemiology, Ebolavirus pathogenicity, Ebolavirus physiology, Global Health, Hemorrhagic Fever, Ebola mortality, Hemorrhagic Fever, Ebola prevention & control, Hemorrhagic Fever, Ebola virology, Humans, Marburg Virus Disease mortality, Marburg Virus Disease prevention & control, Marburg Virus Disease virology, Marburgvirus pathogenicity, Marburgvirus physiology, Survival Analysis, Uganda epidemiology, Biomedical Research organization & administration, Disease Outbreaks, Emergency Medicine organization & administration, Hemorrhagic Fever, Ebola epidemiology, Marburg Virus Disease epidemiology
- Abstract
After more than 28,000 Ebola virus disease cases and at least 11,000 deaths in West Africa during the 2014⁻2016 epidemic, the world remains without a licensed vaccine or therapeutic broadly available and demonstrated to alleviate suffering. This deficiency has been felt acutely in the two, short, following years with two Ebola virus outbreaks in the Democratic Republic of Congo (DRC), and a Marburg virus outbreak in Uganda. Despite billions of U.S. dollars invested in developing medical countermeasures for filoviruses in the antecedent decades, resulting in an array of preventative, diagnostic, and therapeutic products, none are available on commercial shelves. This paper explores why just-in-time research efforts in the field during the West Africa epidemic failed, as well as some recent initiatives to prevent similarly lost opportunities.
- Published
- 2018
- Full Text
- View/download PDF
18. Antibody-Dependent Enhancement of Ebola Virus Infection by Human Antibodies Isolated from Survivors.
- Author
-
Kuzmina NA, Younan P, Gilchuk P, Santos RI, Flyak AI, Ilinykh PA, Huang K, Lubaki NM, Ramanathan P, Crowe JE Jr, and Bukreyev A
- Subjects
- Animals, Antibodies, Monoclonal isolation & purification, Antibodies, Neutralizing isolation & purification, Antibodies, Viral isolation & purification, Ebolavirus drug effects, Ebolavirus genetics, Ebolavirus immunology, Ebolavirus pathogenicity, Epitopes genetics, Epitopes immunology, Gene Expression, Genes, Reporter, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola mortality, Hemorrhagic Fever, Ebola therapy, Humans, Immune Sera chemistry, Immunoglobulin Fc Fragments chemistry, Immunoglobulin Fc Fragments genetics, Marburg Virus Disease immunology, Marburg Virus Disease mortality, Marburg Virus Disease therapy, Marburgvirus drug effects, Marburgvirus genetics, Marburgvirus pathogenicity, Mice, Mice, Inbred BALB C, Monocytes drug effects, Monocytes immunology, Monocytes virology, Primary Cell Culture, Receptors, IgG genetics, Receptors, IgG immunology, Survival Analysis, Survivors, THP-1 Cells, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Antibodies, Monoclonal pharmacology, Antibodies, Neutralizing pharmacology, Antibodies, Viral pharmacology, Antibody-Dependent Enhancement, Hemorrhagic Fever, Ebola virology, Marburg Virus Disease virology
- Abstract
Some monoclonal antibodies (mAbs) recovered from survivors of filovirus infections can protect against infection. It is currently unknown whether natural infection also induces some antibodies with the capacity for antibody-dependent enhancement (ADE). A panel of mAbs obtained from human survivors of filovirus infection caused by Ebola, Bundibugyo, or Marburg viruses was evaluated for their ability to facilitate ADE. ADE was observed readily with all mAbs examined at sub-neutralizing concentrations, and this effect was not restricted to mAbs with a particular epitope specificity, neutralizing capacity, or subclass. Blocking of specific Fcγ receptors reduced but did not abolish ADE that was associated with high-affinity binding antibodies, suggesting that lower-affinity interactions still cause ADE. Mutations of Fc fragments of an mAb that altered its interaction with Fc receptors rendered the antibody partially protective in vivo at a low dose, suggesting that ADE counteracts antibody-mediated protection and facilitates dissemination of filovirus infections., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
19. A bioluminescent imaging mouse model for Marburg virus based on a pseudovirus system.
- Author
-
Zhang L, Li Q, Liu Q, Huang W, Nie J, and Wang Y
- Subjects
- Animals, Disease Models, Animal, Genetic Vectors, Guinea Pigs, Marburg Virus Disease immunology, Marburg Virus Disease prevention & control, Marburgvirus genetics, Marburgvirus immunology, Marburgvirus pathogenicity, Mice, Mice, Inbred BALB C, Neutralization Tests, Virology methods, Luminescent Measurements methods, Marburg Virus Disease virology, Marburgvirus isolation & purification, Viral Vaccines immunology
- Abstract
Marburg virus (MARV) can cause lethal hemorrhagic fever in humans. Handling of MARV is restricted to high-containment biosafety level 4 (BSL-4) facilities, which greatly impedes research into this virus. In this study, a high titer of MARV pseudovirus was generated through optimization of the HIV backbone vectors, the ratio of backbone vector to MARV glycoprotein expression vector, and the transfection reagents. An in vitro neutralization assay and an in vivo bioluminescent imaging mouse model for MARV were developed based on the pseudovirus. Protective serum against MARV was successfully induced in guinea pigs, which showed high neutralization activity in vitro and could also protect Balb/c mice from MARV pseudovirus infection in vivo. This system could be a convenient tool to enable the evaluation of vaccines and therapeutic drugs against MARV in non-BSL-4 laboratories.
- Published
- 2017
- Full Text
- View/download PDF
20. Marburg- and Ebolaviruses: A Look Back and Lessons for the Future.
- Author
-
Klenk HD and Slenczka W
- Subjects
- Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola epidemiology, History, 20th Century, History, 21st Century, Humans, Marburgvirus pathogenicity, Disease Outbreaks history, Hemorrhagic Fever, Ebola history
- Abstract
Since the discovery of Marburg virus 50 years ago, filoviruses have reemerged in the human population more than 40 times. Already the first episode was as dramatic as most of the subsequent ones, but none of them was as devastating as the West-African Ebola virus outbreak in 2013-2015. Although progress toward a better understanding of the viruses is impressive, there is clearly a need to improve and strengthen the measures to detect and control these deadly infections.
- Published
- 2017
- Full Text
- View/download PDF
21. A hamster model for Marburg virus infection accurately recapitulates Marburg hemorrhagic fever.
- Author
-
Marzi A, Banadyga L, Haddock E, Thomas T, Shen K, Horne EJ, Scott DP, Feldmann H, and Ebihara H
- Subjects
- Animals, Blood Coagulation Disorders etiology, Chlorocebus aethiops, Cricetinae, Cytokines genetics, Cytokines metabolism, Disease Models, Animal, Fibrinogen analysis, Hemorrhage etiology, Immunity, Innate, Kaplan-Meier Estimate, Liver pathology, Marburg Virus Disease immunology, Marburg Virus Disease mortality, Marburg Virus Disease virology, Marburgvirus genetics, Marburgvirus isolation & purification, Mutation, Partial Thromboplastin Time, Prothrombin Time, RNA, Viral genetics, RNA, Viral isolation & purification, RNA, Viral metabolism, Spleen pathology, Vero Cells, Marburg Virus Disease pathology, Marburgvirus pathogenicity
- Abstract
Marburg virus (MARV), a close relative of Ebola virus, is the causative agent of a severe human disease known as Marburg hemorrhagic fever (MHF). No licensed vaccine or therapeutic exists to treat MHF, and MARV is therefore classified as a Tier 1 select agent and a category A bioterrorism agent. In order to develop countermeasures against this severe disease, animal models that accurately recapitulate human disease are required. Here we describe the development of a novel, uniformly lethal Syrian golden hamster model of MHF using a hamster-adapted MARV variant Angola. Remarkably, this model displayed almost all of the clinical features of MHF seen in humans and non-human primates, including coagulation abnormalities, hemorrhagic manifestations, petechial rash, and a severely dysregulated immune response. This MHF hamster model represents a powerful tool for further dissecting MARV pathogenesis and accelerating the development of effective medical countermeasures against human MHF.
- Published
- 2016
- Full Text
- View/download PDF
22. Distribution of Marburg virus in Africa: An evolutionary approach.
- Author
-
Zehender G, Sorrentino C, Veo C, Fiaschi L, Gioffrè S, Ebranati E, Tanzi E, Ciccozzi M, Lai A, and Galli M
- Subjects
- Africa epidemiology, Animals, Bayes Theorem, Gene Flow, Humans, Likelihood Functions, Marburg Virus Disease epidemiology, Phylogeny, Phylogeography, Selection, Genetic, Biological Evolution, Marburgvirus genetics, Marburgvirus pathogenicity
- Abstract
The aim of this study was to investigate the origin and geographical dispersion of Marburg virus, the first member of the Filoviridae family to be discovered. Seventy-three complete genome sequences of Marburg virus isolated from animals and humans were retrieved from public databases and analysed using a Bayesian phylogeographical framework. The phylogenetic tree of the Marburg virus data set showed two significant evolutionary lineages: Ravn virus (RAVV) and Marburg virus (MARV). MARV divided into two main clades; clade A included isolates from Uganda (five from the European epidemic in 1967), Kenya (1980) and Angola (from the epidemic of 2004-2005); clade B included most of the isolates obtained during the 1999-2000 epidemic in the Democratic Republic of the Congo (DRC) and a group of Ugandan isolates obtained in 2007-2009. The estimated mean evolutionary rate of the whole genome was 3.3×10(-4) substitutions/site/year (credibility interval 2.0-4.8). The MARV strain had a mean root time of the most recent common ancestor of 177.9years ago (YA) (95% highest posterior density 87-284), thus indicating that it probably originated in the mid-XIX century, whereas the RAVV strain had a later origin dating back to a mean 33.8 YA. The most probable location of the MARV ancestor was Uganda (state posterior probability, spp=0.41), whereas that of the RAVV ancestor was Kenya (spp=0.71). There were significant migration rates from Uganda to the DRC (Bayes Factor, BF=42.0) and in the opposite direction (BF=5.7). Our data suggest that Uganda may have been the cradle of Marburg virus in Africa., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
23. Dimerization Controls Marburg Virus VP24-dependent Modulation of Host Antioxidative Stress Responses.
- Author
-
Johnson B, Li J, Adhikari J, Edwards MR, Zhang H, Schwarz T, Leung DW, Basler CF, Gross ML, and Amarasinghe GK
- Subjects
- Amino Acid Substitution, Binding Sites, Biochemical Phenomena, Cell Line, DNA Mutational Analysis, Epithelial Cells virology, Humans, Marburgvirus immunology, Mass Spectrometry, Models, Molecular, Mutagenesis, Site-Directed, NF-E2-Related Factor 2 metabolism, Protein Binding, Protein Conformation, Sequence Deletion, Viral Proteins chemistry, Virulence Factors chemistry, Virulence Factors metabolism, Host-Pathogen Interactions, Immunity, Innate, Kelch-Like ECH-Associated Protein 1 metabolism, Marburgvirus pathogenicity, Protein Multimerization, Stress, Physiological, Viral Proteins metabolism
- Abstract
Marburg virus (MARV), a member of the Filoviridae family that also includes Ebola virus (EBOV), causes lethal hemorrhagic fever with case fatality rates that have exceeded 50% in some outbreaks. Within an infected cell, there are numerous host-viral interactions that contribute to the outcome of infection. Recent studies identified MARV protein 24 (mVP24) as a modulator of the host antioxidative responses, but the molecular mechanism remains unclear. Using a combination of biochemical and mass spectrometry studies, we show that mVP24 is a dimer in solution that directly binds to the Kelch domain of Kelch-like ECH-associated protein 1 (Keap1) to regulate nuclear factor (erythroid-derived 2)-like 2 (Nrf2). This interaction between Keap1 and mVP24 occurs through the Kelch interaction loop (K-Loop) of mVP24 leading to upregulation of antioxidant response element transcription, which is distinct from other Kelch binders that regulate Nrf2 activity. N-terminal truncations disrupt mVP24 dimerization, allowing monomeric mVP24 to bind Kelch with higher affinity and stimulate higher antioxidative stress response element (ARE) reporter activity. Mass spectrometry-based mapping of the interface revealed overlapping binding sites on Kelch for mVP24 and the Nrf2 proteins. Substitution of conserved cysteines, C209 and C210, to alanine in the mVP24 K-Loop abrogates Kelch binding and ARE activation. Our studies identify a shift in the monomer-dimer equilibrium of MARV VP24, driven by its interaction with Keap1 Kelch domain, as a critical determinant that modulates host responses to pathogenic Marburg viral infections., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Marburg Virus Reverse Genetics Systems.
- Author
-
Schmidt KM and Mühlberger E
- Subjects
- Marburgvirus pathogenicity, Marburgvirus physiology, Marburgvirus genetics, Reverse Genetics methods, Virology methods
- Abstract
The highly pathogenic Marburg virus (MARV) is a member of the Filoviridae family and belongs to the group of nonsegmented negative-strand RNA viruses. Reverse genetics systems established for MARV have been used to study various aspects of the viral replication cycle, analyze host responses, image viral infection, and screen for antivirals. This article provides an overview of the currently established MARV reverse genetic systems based on minigenomes, infectious virus-like particles and full-length clones, and the research that has been conducted using these systems.
- Published
- 2016
- Full Text
- View/download PDF
25. Comparison of the Pathogenesis of the Angola and Ravn Strains of Marburg Virus in the Outbred Guinea Pig Model.
- Author
-
Cross RW, Fenton KA, Geisbert JB, Ebihara H, Mire CE, and Geisbert TW
- Subjects
- Angola, Animals, Blood Coagulation physiology, Cytokines metabolism, Female, Genetic Linkage genetics, Genetic Variation genetics, Guinea Pigs, Marburg Virus Disease metabolism, Marburgvirus genetics, Nitric Oxide metabolism, Viremia metabolism, Marburg Virus Disease virology, Marburgvirus pathogenicity, Virulence genetics
- Abstract
Background: Phylogenetic comparisons of known Marburg virus (MARV) strains reveal 2 distinct genetic lineages: Ravn and the Lake Victoria Marburg complex (eg, Musoke, Popp, and Angola strains). Nucleotide variances of >20% between Ravn and other MARV genomes suggest that differing virulence between lineages may accompany this genetic divergence. To date, there exists limited systematic experimental evidence of pathogenic differences between MARV strains., Methods: Uniformly lethal outbred guinea pig models of MARV-Angola (MARV-Ang) and MARV-Ravn (MARV-Rav) were developed by serial adaptation. Changes in genomic sequence, weight, temperature, histopathologic findings, immunohistochemical findings, hematologic profiles, circulating biochemical enzyme levels, coagulation parameters, viremia levels, cytokine levels, eicanosoid levels, and nitric oxide production were compared between strains., Results: MARV-Rav infection resulted in delayed increases in circulating inflammatory and prothrombotic elements, notably lower viremia levels, less severe histologic alterations, and a delay in mean time to death, compared with MARV-Ang infection. Both strains produced more marked coagulation abnormalities than previously seen in MARV-infected mice or inbred guinea pigs., Conclusions: Although both strains exhibit great similarity to pathogenic markers of human and nonhuman primate MARV infection, these data highlight several key differences in pathogenicity that may serve to guide the choice of strain and model used for development of vaccines or therapeutics for Marburg hemorrhagic fever., (Published by Oxford University Press on behalf of the Infectious Diseases Society of America 2015. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2015
- Full Text
- View/download PDF
26. Amino Acid Residue at Position 79 of Marburg Virus VP40 Confers Interferon Antagonism in Mouse Cells.
- Author
-
Feagins AR and Basler CF
- Subjects
- Animals, Cell Line, HEK293 Cells, Humans, Interferon-alpha metabolism, Interferon-beta metabolism, Janus Kinase 1 metabolism, Marburg Virus Disease metabolism, Marburg Virus Disease virology, Mice, STAT1 Transcription Factor metabolism, STAT2 Transcription Factor metabolism, Amino Acids metabolism, Marburgvirus metabolism, Marburgvirus pathogenicity, Viral Matrix Proteins metabolism
- Abstract
Marburg viruses (MARVs) cause highly lethal infections in humans and nonhuman primates. Mice are not generally susceptible to MARV infection; however, if the strain is first adapted to mice through serial passaging, it becomes able to cause disease in this animal. A previous study correlated changes accrued during mouse adaptation in the VP40 gene of a MARV strain known as Ravn virus (RAVV) with an increased capacity to inhibit interferon (IFN) signaling in mouse cell lines. The MARV strain Ci67, which belongs to a different phylogenetic clade than RAVV, has also been adapted to mice and in the process the Ci67 VP40 acquired a different collection of genetic changes than did RAVV VP40. Here, we demonstrate that the mouse-adapted Ci67 VP40 more potently antagonizes IFN-α/β-induced STAT1 and STAT2 tyrosine phosphorylation, gene expression, and antiviral activity in both mouse and human cell lines, compared with the parental Ci67 VP40. Ci67 VP40 is also demonstrated to target the activation of kinase Jak1. A single change at VP40 residue 79 was found to be sufficient for the increased VP40 IFN antagonism. These data argue that VP40 IFN-antagonist activity plays a key role in MARV pathogenesis in mice., (© The Author 2015. Published by Oxford University Press on behalf of the Infectious Diseases Society of America. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
27. Temporal Characterization of Marburg Virus Angola Infection following Aerosol Challenge in Rhesus Macaques.
- Author
-
Lin KL, Twenhafel NA, Connor JH, Cashman KA, Shamblin JD, Donnelly GC, Esham HL, Wlazlowski CB, Johnson JC, Honko AN, Botto MA, Yen J, Hensley LE, and Goff AJ
- Subjects
- Aerosols, Animals, Disease Progression, Immunohistochemistry, Longitudinal Studies, Macaca mulatta, Marburg Virus Disease blood, Time Factors, Viral Load, Cytokines blood, Host-Pathogen Interactions, Marburg Virus Disease physiopathology, Marburgvirus pathogenicity
- Abstract
Unlabelled: Marburg virus (MARV) infection is a lethal hemorrhagic fever for which no licensed vaccines or therapeutics are available. Development of appropriate medical countermeasures requires a thorough understanding of the interaction between the host and the pathogen and the resulting disease course. In this study, 15 rhesus macaques were sequentially sacrificed following aerosol exposure to the MARV variant Angola, with longitudinal changes in physiology, immunology, and histopathology used to assess disease progression. Immunohistochemical evidence of infection and resulting histopathological changes were identified as early as day 3 postexposure (p.e.). The appearance of fever in infected animals coincided with the detection of serum viremia and plasma viral genomes on day 4 p.e. High (>10(7) PFU/ml) viral loads were detected in all major organs (lung, liver, spleen, kidney, brain, etc.) beginning day 6 p.e. Clinical pathology findings included coagulopathy, leukocytosis, and profound liver destruction as indicated by elevated liver transaminases, azotemia, and hypoalbuminemia. Altered cytokine expression in response to infection included early increases in Th2 cytokines such as interleukin 10 (IL-10) and IL-5 and late-stage increases in Th1 cytokines such as IL-2, IL-15, and granulocyte-macrophage colony-stimulating factor (GM-CSF). This study provides a longitudinal examination of clinical disease of aerosol MARV Angola infection in the rhesus macaque model., Importance: In this study, we carefully analyzed the timeline of Marburg virus infection in nonhuman primates in order to provide a well-characterized model of disease progression following aerosol exposure., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
28. Lack of Marburg Virus Transmission From Experimentally Infected to Susceptible In-Contact Egyptian Fruit Bats.
- Author
-
Paweska JT, Jansen van Vuren P, Fenton KA, Graves K, Grobbelaar AA, Moolla N, Leman P, Weyer J, Storm N, McCulloch SD, Scott TP, Markotter W, Odendaal L, Clift SJ, Geisbert TW, Hale MJ, and Kemp A
- Subjects
- Animals, Disease Outbreaks, Disease Susceptibility blood, Disease Susceptibility immunology, Female, Humans, Male, Marburg Virus Disease immunology, Marburg Virus Disease virology, Marburgvirus genetics, Marburgvirus immunology, Virus Replication genetics, Chiroptera virology, Disease Susceptibility virology, Marburg Virus Disease transmission, Marburgvirus pathogenicity
- Abstract
Egyptian fruit bats (Rousettus aegyptiacus) were inoculated subcutaneously (n = 22) with Marburg virus (MARV). No deaths, overt signs of morbidity, or gross lesions was identified, but microscopic pathological changes were seen in the liver of infected bats. The virus was detected in 15 different tissues and plasma but only sporadically in mucosal swab samples, urine, and fecal samples. Neither seroconversion nor viremia could be demonstrated in any of the in-contact susceptible bats (n = 14) up to 42 days after exposure to infected bats. In bats rechallenged (n = 4) on day 48 after infection, there was no viremia, and the virus could not be isolated from any of the tissues tested. This study confirmed that infection profiles are consistent with MARV replication in a reservoir host but failed to demonstrate MARV transmission through direct physical contact or indirectly via air. Bats develop strong protective immunity after infection with MARV., (© The Author 2015. Published by Oxford University Press on behalf of the Infectious Diseases Society of America. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
29. Inhibition of Ebola and Marburg Virus Entry by G Protein-Coupled Receptor Antagonists.
- Author
-
Cheng H, Lear-Rooney CM, Johansen L, Varhegyi E, Chen ZW, Olinger GG, and Rong L
- Subjects
- Animals, Antiviral Agents pharmacology, Benztropine pharmacology, Cell Line, Chlorocebus aethiops, Cyproheptadine pharmacology, Ebolavirus pathogenicity, HEK293 Cells, Hemorrhagic Fever, Ebola drug therapy, Heparin pharmacology, High-Throughput Screening Assays, Humans, Macrolides pharmacology, Marburg Virus Disease drug therapy, Marburgvirus pathogenicity, Receptors, G-Protein-Coupled physiology, Small Molecule Libraries, Vero Cells, Zidovudine pharmacology, Ebolavirus drug effects, Ebolavirus physiology, Marburgvirus drug effects, Marburgvirus physiology, Receptors, G-Protein-Coupled antagonists & inhibitors, Virus Internalization drug effects
- Abstract
Unlabelled: Filoviruses, consisting of Ebola virus (EBOV) and Marburg virus (MARV), are among the most lethal infectious threats to mankind. Infections by these viruses can cause severe hemorrhagic fevers in humans and nonhuman primates with high mortality rates. Since there is currently no vaccine or antiviral therapy approved for humans, there is an urgent need to develop prophylactic and therapeutic options for use during filoviral outbreaks and bioterrorist attacks. One of the ideal targets against filoviral infection and diseases is at the entry step, which is mediated by the filoviral glycoprotein (GP). In this report, we screened a chemical library of small molecules and identified numerous inhibitors, which are known G protein-coupled receptor (GPCR) antagonists targeting different GPCRs, including histamine receptors, 5-HT (serotonin) receptors, muscarinic acetylcholine receptor, and adrenergic receptor. These inhibitors can effectively block replication of both infectious EBOV and MARV, indicating a broad antiviral activity of the GPCR antagonists. The time-of-addition experiment and microscopic studies suggest that GPCR antagonists block filoviral entry at a step following the initial attachment but prior to viral/cell membrane fusion. These results strongly suggest that GPCRs play a critical role in filoviral entry and GPCR antagonists can be developed as an effective anti-EBOV/MARV therapy., Importance: Infection of Ebola virus and Marburg virus can cause severe illness in humans with a high mortality rate, and currently there is no FDA-approved vaccine or therapeutic treatment available. The 2013-2015 epidemic in West Africa underscores a lack of our understanding in the infection and pathogenesis of these viruses and the urgency of drug discovery and development. In this study, we have identified numerous inhibitors that are known G protein-coupled receptor (GPCR) antagonists targeting different GPCRs. These inhibitors can effectively block replication of both infectious EBOV and MARV, indicating a broad antiviral activity of the GPCR antagonists. Our results strongly suggest that GPCRs play a critical role in filoviral entry and GPCR antagonists can be developed as an effective anti-EBOV/MARV therapy., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
30. Role of EXT1 and Glycosaminoglycans in the Early Stage of Filovirus Entry.
- Author
-
O'Hearn A, Wang M, Cheng H, Lear-Rooney CM, Koning K, Rumschlag-Booms E, Varhegyi E, Olinger G, and Rong L
- Subjects
- Animals, Cell Line, Ebolavirus pathogenicity, Ebolavirus physiology, Filoviridae pathogenicity, Filoviridae Infections etiology, Filoviridae Infections virology, Gene Knockdown Techniques, HEK293 Cells, Heparin physiology, Heparitin Sulfate biosynthesis, Heparitin Sulfate deficiency, Host-Pathogen Interactions, Humans, Marburgvirus pathogenicity, Marburgvirus physiology, Mice, N-Acetylglucosaminyltransferases antagonists & inhibitors, N-Acetylglucosaminyltransferases genetics, Receptors, Virus physiology, Viral Proteins physiology, Virulence, Filoviridae physiology, Glycosaminoglycans physiology, N-Acetylglucosaminyltransferases physiology, Virus Internalization
- Abstract
Unlabelled: Filoviruses, including both Ebola virus (EBOV) and Marburg virus (MARV), can infect humans and other animals, causing hemorrhagic fever with a high mortality rate. Entry of these viruses into the host is mediated by a single filoviral glycoprotein (GP). GP is composed of two subunits: GP1, which is responsible for attachment and binding to receptor(s) on susceptible cells, and GP2, which mediates viral and cell membrane fusion. Although numerous host factors have been implicated in the entry process, the initial attachment receptor(s) has not been well defined. In this report, we demonstrate that exostosin 1 (EXT1), which is involved in biosynthesis of heparan sulfate (HS), plays a role in filovirus entry. Expression knockdown of EXT1 by small interfering RNAs (siRNAs) impairs GP-mediated pseudoviral entry and that of infectious EBOV and MARV in tissue cultured cells. Furthermore, HS, heparin, and other related glycosaminoglycans (GAGs), to different extents, can bind to and block GP-mediated viral entry and that of infectious filoviruses. These results strongly suggest that HS and other related GAGs are attachment receptors that are utilized by filoviruses for entry and infection. These GAGs may have therapeutic potential in treating EBOV- and MARV-infected patients., Importance: Infection by Ebola virus and Marburg virus can cause severe illness in humans, with a high mortality rate, and currently there is no FDA-approved vaccine or therapeutic treatment available. The ongoing 2014 outbreak in West Africa underscores a lack of our understanding in the infection and pathogenesis of these viruses and the urgency of drug discovery and development. In this study, we provide several pieces of evidence that demonstrate that heparan sulfate and other closely related glycosaminoglycans are the molecules that are used by filoviruses for initial attachment. Furthermore, we demonstrate that these glycosaminoglycans can block entry of and infection by filoviruses. Thus, this work provides mechanistic insights on the early step of filoviral infection and suggests a possible therapeutic option for diseases caused by filovirus infection., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
31. Ebola and Marburg haemorrhagic fever.
- Author
-
Rougeron V, Feldmann H, Grard G, Becker S, and Leroy EM
- Subjects
- Africa epidemiology, Animals, Disease Outbreaks, Ebolavirus pathogenicity, Humans, Marburgvirus pathogenicity, Prevalence, Hemorrhagic Fever, Ebola diagnosis, Hemorrhagic Fever, Ebola epidemiology, Hemorrhagic Fever, Ebola prevention & control, Hemorrhagic Fever, Ebola therapy, Marburg Virus Disease diagnosis, Marburg Virus Disease epidemiology, Marburg Virus Disease prevention & control, Marburg Virus Disease therapy
- Abstract
Ebolaviruses and Marburgviruses (family Filoviridae) are among the most virulent pathogens for humans and great apes causing severe haemorrhagic fever and death within a matter of days. This group of viruses is characterized by a linear, non-segmented, single-stranded RNA genome of negative polarity. The overall burden of filovirus infections is minimal and negligible compared to the devastation caused by malnutrition and other infectious diseases prevalent in Africa such as malaria, dengue or tuberculosis. In this paper, we review the knowledge gained on the eco/epidemiology, the pathogenesis and the disease control measures for Marburg and Ebola viruses developed over the last 15 years. The overall progress is promising given the little attention that these pathogen have achieved in the past; however, more is to come over the next decade given the more recent interest in these pathogens as potential public and animal health concerns. Licensing of therapeutic and prophylactic options may be achievable over the next 5-10 years., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. Tissue and cellular tropism, pathology and pathogenesis of Ebola and Marburg viruses.
- Author
-
Martines RB, Ng DL, Greer PW, Rollin PE, and Zaki SR
- Subjects
- Animals, Biopsy, Ebolavirus genetics, Ebolavirus immunology, Ebolavirus isolation & purification, Hemorrhagic Fever, Ebola diagnosis, Hemorrhagic Fever, Ebola epidemiology, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola transmission, Host-Pathogen Interactions, Humans, Marburg Virus Disease diagnosis, Marburg Virus Disease epidemiology, Marburg Virus Disease immunology, Marburg Virus Disease transmission, Marburgvirus genetics, Marburgvirus immunology, Marburgvirus isolation & purification, Pathology, Molecular methods, Predictive Value of Tests, Prognosis, Risk Factors, Virology methods, Virulence, Virus Internalization, Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola pathology, Hemorrhagic Fever, Ebola virology, Marburg Virus Disease pathology, Marburg Virus Disease virology, Marburgvirus pathogenicity, Viral Tropism
- 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., (Copyright © 2014 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
33. Durability of a vesicular stomatitis virus-based marburg virus vaccine in nonhuman primates.
- Author
-
Mire CE, Geisbert JB, Agans KN, Satterfield BA, Versteeg KM, Fritz EA, Feldmann H, Hensley LE, and Geisbert TW
- Subjects
- Animals, Ebola Vaccines therapeutic use, Female, Immunity, Cellular physiology, Immunity, Humoral physiology, Macaca, Male, Marburg Virus Disease immunology, Marburg Virus Disease prevention & control, Marburg Virus Disease virology, Marburgvirus pathogenicity, Primates, Vesicular Stomatitis virology, Viremia immunology, Viremia prevention & control, Viremia virology, Marburgvirus immunology, Vesicular Stomatitis immunology, Vesicular Stomatitis prevention & control
- Abstract
The filoviruses, Marburg virus (MARV) and Ebola virus, causes severe hemorrhagic fever with high mortality in humans and nonhuman primates. A promising filovirus vaccine under development is based on a recombinant vesicular stomatitis virus (rVSV) that expresses individual filovirus glycoproteins (GPs) in place of the VSV glycoprotein (G). These vaccines have shown 100% efficacy against filovirus infection in nonhuman primates when challenge occurs 28-35 days after a single injection immunization. Here, we examined the ability of a rVSV MARV-GP vaccine to provide protection when challenge occurs more than a year after vaccination. Cynomolgus macaques were immunized with rVSV-MARV-GP and challenged with MARV approximately 14 months after vaccination. Immunization resulted in the vaccine cohort of six animals having anti-MARV GP IgG throughout the pre-challenge period. Following MARV challenge none of the vaccinated animals showed any signs of clinical disease or viremia and all were completely protected from MARV infection. Two unvaccinated control animals exhibited signs consistent with MARV infection and both succumbed. Importantly, these data are the first to show 100% protective efficacy against any high dose filovirus challenge beyond 8 weeks after final vaccination. These findings demonstrate the durability of VSV-based filovirus vaccines.
- Published
- 2014
- Full Text
- View/download PDF
34. Induction of broad cytotoxic T cells by protective DNA vaccination against Marburg and Ebola.
- Author
-
Shedlock DJ, Aviles J, Talbott KT, Wong G, Wu SJ, Villarreal DO, Myles DJ, Croyle MA, Yan J, Kobinger GP, and Weiner DB
- Subjects
- Animals, Cell Line, Enzyme-Linked Immunosorbent Assay, Female, Flow Cytometry, Humans, Immunoblotting, Marburgvirus immunology, Marburgvirus pathogenicity, Mice, Inbred C57BL, Vaccines, DNA therapeutic use, Viral Vaccines immunology, Viral Vaccines therapeutic use, Marburg Virus Disease immunology, Marburg Virus Disease prevention & control, T-Lymphocytes, Cytotoxic immunology, Vaccines, DNA immunology
- Abstract
Marburg and Ebola hemorrhagic fevers have been described as the most virulent viral diseases known to man due to associative lethality rates of up to 90%. Death can occur within days to weeks of exposure and there is currently no licensed vaccine or therapeutic. Recent evidence suggests an important role for antiviral T cells in conferring protection, but little detailed analysis of this response as driven by a protective vaccine has been reported. We developed a synthetic polyvalent-filovirus DNA vaccine against Marburg marburgvirus (MARV), Zaire ebolavirus (ZEBOV), and Sudan ebolavirus (SUDV). Preclinical efficacy studies were performed in guinea pigs and mice using rodent-adapted viruses, whereas murine T-cell responses were extensively analyzed using a novel modified assay described herein. Vaccination was highly potent, elicited robust neutralizing antibodies, and completely protected against MARV and ZEBOV challenge. Comprehensive T-cell analysis revealed cytotoxic T lymphocytes (CTLs) of great magnitude, epitopic breadth, and Th1-type marker expression. This model provides an important preclinical tool for studying protective immune correlates that could be applied to existing platforms. Data herein support further evaluation of this enhanced gene-based approach in nonhuman primate studies for in depth analyses of T-cell epitopes in understanding protective efficacy.
- Published
- 2013
- Full Text
- View/download PDF
35. Small molecule inhibitors of ER α-glucosidases are active against multiple hemorrhagic fever viruses.
- Author
-
Chang J, Warren TK, Zhao X, Gill T, Guo F, Wang L, Comunale MA, Du Y, Alonzi DS, Yu W, Ye H, Liu F, Guo JT, Mehta A, Cuconati A, Butters TD, Bavari S, Xu X, and Block TM
- Subjects
- Animals, Antiviral Agents pharmacokinetics, Dengue drug therapy, Dogs, Drug Evaluation, Preclinical, Ebolavirus drug effects, Ebolavirus pathogenicity, Endoplasmic Reticulum enzymology, HEK293 Cells, Humans, Imino Sugars pharmacokinetics, Marburg Virus Disease drug therapy, Marburgvirus drug effects, Marburgvirus pathogenicity, Mice, Mice, Inbred BALB C, Rats, Rats, Sprague-Dawley, Structure-Activity Relationship, alpha-Glucosidases, Antiviral Agents pharmacology, Glycoside Hydrolase Inhibitors, Hemorrhagic Fever, Ebola drug therapy, Imino Sugars pharmacology
- Abstract
Host cellular endoplasmic reticulum α-glucosidases I and II are essential for the maturation of viral glycosylated envelope proteins that use the calnexin mediated folding pathway. Inhibition of these glycan processing enzymes leads to the misfolding and degradation of these viral glycoproteins and subsequent reduction in virion secretion. We previously reported that, CM-10-18, an imino sugar α-glucosidase inhibitor, efficiently protected the lethality of dengue virus infection of mice. In the current study, through an extensive structure-activity relationship study, we have identified three CM-10-18 derivatives that demonstrated superior in vitro antiviral activity against representative viruses from four viral families causing hemorrhagic fever. Moreover, the three novel imino sugars significantly reduced the mortality of two of the most pathogenic hemorrhagic fever viruses, Marburg virus and Ebola virus, in mice. Our study thus proves the concept that imino sugars are promising drug candidates for the management of viral hemorrhagic fever caused by variety of viruses., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
36. Experimental respiratory Marburg virus haemorrhagic fever infection in the common marmoset (Callithrix jacchus).
- Author
-
Smither SJ, Nelson M, Eastaugh L, Laws TR, Taylor C, Smith SA, Salguero FJ, and Lever MS
- Subjects
- Animals, Cytokines metabolism, Disease Susceptibility, Female, Humans, Kidney pathology, Kidney virology, Liver pathology, Liver virology, Lung pathology, Lung virology, Male, Marburg Virus Disease virology, Marburgvirus isolation & purification, Monkey Diseases virology, Spleen pathology, Spleen virology, Callithrix, Disease Models, Animal, Inhalation Exposure, Marburg Virus Disease pathology, Marburgvirus pathogenicity, Monkey Diseases pathology
- Abstract
Marburg virus causes a highly infectious and lethal haemorrhagic fever in primates and may be exploited as a potential biothreat pathogen. To combat the infection and threat of Marburg haemorrhagic fever, there is a need to develop and license appropriate medical countermeasures. To determine whether the common marmoset (Callithrix jacchus) would be an appropriate model to assess therapies against Marburg haemorrhagic fever, initial susceptibility, lethality and pathogenesis studies were performed. Low doses of virus, between 4 and 28 TCID50 , were sufficient to cause a lethal, reproducible infection. Animals became febrile between days 5 and 6, maintaining a high fever before succumbing to disease between 8 and 11 days postchallenge. Typical signs of Marburg virus infection were observed including haemorrhaging and a transient rash. In pathogenesis studies, virus was isolated from the animals' lungs from day 3 postchallenge and from the liver, spleen and blood from day 5 postchallenge. Early signs of histopathology were apparent in the kidney and liver from day 3. The most striking features were observed in animals exhibiting severe clinical signs, which included high viral titres in all organs, with the highest levels in the blood, increased levels in liver function enzymes and blood clotting times, decreased levels in platelets, multifocal moderate-to-severe hepatitis and perivascular oedema., (© 2013 Crown copyright. International Journal of Experimental Pathology © 2013 International Journal of Experimental Pathology.)
- Published
- 2013
- Full Text
- View/download PDF
37. Structural basis for Marburg virus VP35-mediated immune evasion mechanisms.
- Author
-
Ramanan P, Edwards MR, Shabman RS, Leung DW, Endlich-Frazier AC, Borek DM, Otwinowski Z, Liu G, Huh J, Basler CF, and Amarasinghe GK
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Crystallography, X-Ray, HEK293 Cells, Host-Pathogen Interactions, Humans, I-kappa B Kinase antagonists & inhibitors, Immunity, Innate, Interferon Type I antagonists & inhibitors, Marburg Virus Disease etiology, Marburg Virus Disease immunology, Marburg Virus Disease virology, Marburgvirus chemistry, Models, Biological, Models, Molecular, Molecular Sequence Data, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Structure, Tertiary, RNA chemistry, RNA genetics, RNA metabolism, Sequence Homology, Amino Acid, Virulence immunology, Marburgvirus immunology, Marburgvirus pathogenicity, Viral Regulatory and Accessory Proteins chemistry, Viral Regulatory and Accessory Proteins immunology
- Abstract
Filoviruses, marburgvirus (MARV) and ebolavirus (EBOV), are causative agents of highly lethal hemorrhagic fever in humans. MARV and EBOV share a common genome organization but show important differences in replication complex formation, cell entry, host tropism, transcriptional regulation, and immune evasion. Multifunctional filoviral viral protein (VP) 35 proteins inhibit innate immune responses. Recent studies suggest double-stranded (ds)RNA sequestration is a potential mechanism that allows EBOV VP35 to antagonize retinoic-acid inducible gene-I (RIG-I) like receptors (RLRs) that are activated by viral pathogen-associated molecular patterns (PAMPs), such as double-strandedness and dsRNA blunt ends. Here, we show that MARV VP35 can inhibit IFN production at multiple steps in the signaling pathways downstream of RLRs. The crystal structure of MARV VP35 IID in complex with 18-bp dsRNA reveals that despite the similar protein fold as EBOV VP35 IID, MARV VP35 IID interacts with the dsRNA backbone and not with blunt ends. Functional studies show that MARV VP35 can inhibit dsRNA-dependent RLR activation and interferon (IFN) regulatory factor 3 (IRF3) phosphorylation by IFN kinases TRAF family member-associated NFkb activator (TANK) binding kinase-1 (TBK-1) and IFN kB kinase e (IKKe) in cell-based studies. We also show that MARV VP35 can only inhibit RIG-I and melanoma differentiation associated gene 5 (MDA5) activation by double strandedness of RNA PAMPs (coating backbone) but is unable to inhibit activation of RLRs by dsRNA blunt ends (end capping). In contrast, EBOV VP35 can inhibit activation by both PAMPs. Insights on differential PAMP recognition and inhibition of IFN induction by a similar filoviral VP35 fold, as shown here, reveal the structural and functional plasticity of a highly conserved virulence factor.
- Published
- 2012
- Full Text
- View/download PDF
38. A multiagent filovirus DNA vaccine delivered by intramuscular electroporation completely protects mice from ebola and Marburg virus challenge.
- Author
-
Grant-Klein RJ, Van Deusen NM, Badger CV, Hannaman D, Dupuy LC, and Schmaljohn CS
- Subjects
- Animals, Ebolavirus immunology, Ebolavirus pathogenicity, Female, Hemorrhagic Fever, Ebola immunology, Marburg Virus Disease immunology, Marburgvirus immunology, Marburgvirus pathogenicity, Mice, Mice, Inbred BALB C, Electroporation methods, Hemorrhagic Fever, Ebola prevention & control, Marburg Virus Disease prevention & control, Muscles metabolism, Vaccines, DNA administration & dosage, Vaccines, DNA therapeutic use
- Abstract
We evaluated the immunogenicity and protective efficacy of DNA vaccines expressing the codon-optimized envelope glycoprotein genes of Zaire ebolavirus, Sudan ebolavirus, and Marburg marburgvirus (Musoke and Ravn). Intramuscular or intradermal delivery of the vaccines in BALB/c mice was performed using the TriGrid™ electroporation device. Mice that received DNA vaccines against the individual viruses developed robust glycoprotein-specific antibody titers as determined by ELISA and survived lethal viral challenge with no display of clinical signs of infection. Survival curve analysis revealed there was a statistically significant increase in survival compared to the control groups for both the Ebola and Ravn virus challenges. These data suggest that further analysis of the immune responses generated in the mice and additional protection studies in nonhuman primates are warranted.
- Published
- 2012
- Full Text
- View/download PDF
39. The Baboon (Papio spp.) as a model of human Ebola virus infection.
- Author
-
Perry DL, Bollinger L, and White GL
- Subjects
- Animals, Base Sequence, Blood Coagulation Factors metabolism, Fibrin metabolism, Hemorrhagic Fever, Ebola metabolism, Hemorrhagic Fever, Ebola virology, Humans, Lymphatic Diseases pathology, Lymphatic Diseases virology, Marburg Virus Disease metabolism, Marburg Virus Disease virology, Marburgvirus pathogenicity, Necrosis pathology, Necrosis virology, Sequence Homology, Nucleic Acid, Species Specificity, Thrombocytopenia pathology, Thrombocytopenia virology, Disease Models, Animal, Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola pathology, Marburg Virus Disease pathology, Papio virology
- Abstract
Baboons are susceptible to natural Ebola virus (EBOV) infection and share 96% genetic homology with humans. Despite these characteristics, baboons have rarely been utilized as experimental models of human EBOV infection to evaluate the efficacy of prophylactics and therapeutics in the United States. This review will summarize what is known about the pathogenesis of EBOV infection in baboons compared to EBOV infection in humans and other Old World nonhuman primates. In addition, we will discuss how closely the baboon model recapitulates human EBOV infection. We will also review some of the housing requirements and behavioral attributes of baboons compared to other Old World nonhuman primates. Due to the lack of data available on the pathogenesis of Marburg virus (MARV) infection in baboons, discussion of the pathogenesis of MARV infection in baboons will be limited.
- Published
- 2012
- Full Text
- View/download PDF
40. Forty-five years of Marburg virus research.
- Author
-
Brauburger K, Hume AJ, Mühlberger E, and Olejnik J
- Subjects
- Animals, Chiroptera virology, Disease Transmission, Infectious prevention & control, Disease Vectors, Humans, Marburg Virus Disease epidemiology, Marburg Virus Disease pathology, Marburgvirus genetics, Phylogeny, Time Factors, Viral Proteins genetics, Virus Internalization, Disease Outbreaks prevention & control, Genome, Viral, Marburg Virus Disease virology, Marburgvirus pathogenicity
- Abstract
In 1967, the first reported filovirus hemorrhagic fever outbreak took place in Germany and the former Yugoslavia. The causative agent that was identified during this outbreak, Marburg virus, is one of the most deadly human pathogens. This article provides a comprehensive overview of our current knowledge about Marburg virus disease ranging from ecology to pathogenesis and molecular biology.
- Published
- 2012
- Full Text
- View/download PDF
41. Mouse models for filovirus infections.
- Author
-
Bradfute SB, Warfield KL, and Bray M
- Subjects
- Animals, Mice, Disease Models, Animal, Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola pathology, Hemorrhagic Fever, Ebola virology, Marburg Virus Disease pathology, Marburg Virus Disease virology, Marburgvirus pathogenicity
- Abstract
The filoviruses marburg- and ebolaviruses can cause severe hemorrhagic fever (HF) in humans and nonhuman primates. Because many cases have occurred in geographical areas lacking a medical research infrastructure, most studies of the pathogenesis of filoviral HF, and all efforts to develop drugs and vaccines, have been carried out in biocontainment laboratories in non-endemic countries, using nonhuman primates (NHPs), guinea pigs and mice as animal models. NHPs appear to closely mirror filoviral HF in humans (based on limited clinical data), but only small numbers may be used in carefully regulated experiments; much research is therefore done in rodents. Because of their availability in large numbers and the existence of a wealth of reagents for biochemical and immunological testing, mice have become the preferred small animal model for filovirus research. Since the first experiments following the initial 1967 marburgvirus outbreak, wild-type or mouse-adapted viruses have been tested in immunocompetent or immunodeficient mice. In this paper, we review how these types of studies have been used to investigate the pathogenesis of filoviral disease, identify immune responses to infection and evaluate antiviral drugs and vaccines. We also discuss the strengths and weaknesses of murine models for filovirus research, and identify important questions for further study.
- Published
- 2012
- Full Text
- View/download PDF
42. Lethality and pathogenesis of airborne infection with filoviruses in A129 α/β -/- interferon receptor-deficient mice.
- Author
-
Lever MS, Piercy TJ, Steward JA, Eastaugh L, Smither SJ, Taylor C, Salguero FJ, and Phillpotts RJ
- Subjects
- Animals, Ebolavirus pathogenicity, Female, Filoviridae classification, Filoviridae Infections virology, Hemorrhagic Fever, Ebola mortality, Hemorrhagic Fever, Ebola physiopathology, Hemorrhagic Fever, Ebola virology, Humans, Injections, Intraperitoneal, Male, Marburg Virus Disease mortality, Marburg Virus Disease physiopathology, Marburg Virus Disease virology, Marburgvirus pathogenicity, Mice, Mice, Knockout, Receptor, Interferon alpha-beta deficiency, Virulence, Aerosols, Disease Models, Animal, Filoviridae pathogenicity, Filoviridae Infections mortality, Filoviridae Infections physiopathology, Receptor, Interferon alpha-beta genetics
- Abstract
Normal immunocompetent mice are not susceptible to non-adapted filoviruses. There are therefore two strategies available to establish a murine model of filovirus infection: adaptation of the virus to the host or the use of genetically modified mice that are susceptible to the virus. A number of knockout (KO) strains of mice with defects in either their adaptive or innate immunity are susceptible to non-adapted filoviruses. In this study, A129 α/β -/- interferon receptor-deficient KO mice, strain A129 IFN-α/β -/-, were used to determine the lethality of a range of filoviruses, including Lake Victoria marburgvirus (MARV), Zaire ebolavirus (ZEBOV), Sudan ebolavirus (SEBOV), Reston ebolavirus (REBOV) and Côte d'Ivoire ebolavirus (CIEBOV), administered by using intraperitoneal (IP) or aerosol routes of infection. One hundred percent mortality was observed in all groups of KO mice that were administered with a range of challenge doses of MARV and ZEBOV by either IP or aerosol routes. Mean time to death for both routes was dose-dependent and ranged from 5.4 to 7.4 days in the IP injection challenge, and from 10.2 to 13 days in the aerosol challenge. The lethal dose (50 % tissue culture infective dose, TCID(50)) of ZEBOV for KO mice was <1 TCID(50) ml(-1) when administered by either the IP or aerosol route of infection; for MARV the lethal dose was <1 TCID(50) ml(-1) by the IP route of infection and <10 TCID(50) ml(-1) by the aerosol route. In contrast, there was no mortality after infection with SEBOV or REBOV by either IP or aerosol routes of infection; all the mice lost weight (~15 % loss of group mean body weight with SEBOV and ~7 % with REBOV) but recovered to their original weights by day 14 post-challenge. There was no mortality in mice administered with CIEBOV via the IP route of infection and no clinical signs of infection were observed. The progression of disease was faster following infection with ZEBOV than with MARV but ultimately both viruses caused widespread infection with high titres of the infectious viruses in multiple organs. Histopathological observations were consistent with other animal models and showed widespread organ damage. This study suggests that MARV and ZEBOV are more virulent when administered via the IP route rather than by aerosol infection, although both are highly virulent by either route. The KO mouse may provide a useful model to test potential antiviral therapeutics against wild-type filoviruses.
- Published
- 2012
- Full Text
- View/download PDF
43. Recombinant vesicular stomatitis virus vaccine vectors expressing filovirus glycoproteins lack neurovirulence in nonhuman primates.
- Author
-
Mire CE, Miller AD, Carville A, Westmoreland SV, Geisbert JB, Mansfield KG, Feldmann H, Hensley LE, and Geisbert TW
- Subjects
- Animals, Ebolavirus immunology, Genetic Vectors, Glycoproteins genetics, Macaca, Male, Marburgvirus immunology, Primate Diseases pathology, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated adverse effects, Vaccines, Attenuated genetics, Vaccines, Attenuated immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic adverse effects, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Vesiculovirus genetics, Viral Proteins genetics, Viral Vaccines administration & dosage, Viral Vaccines genetics, Viral Vaccines immunology, Ebolavirus pathogenicity, Encephalitis, Viral pathology, Glycoproteins immunology, Marburgvirus pathogenicity, Vesiculovirus pathogenicity, Viral Proteins immunology, Viral Vaccines adverse effects
- Abstract
The filoviruses, Marburg virus and Ebola virus, cause severe hemorrhagic fever with high mortality in humans and nonhuman primates. Among the most promising filovirus vaccines under development is a system based on recombinant vesicular stomatitis virus (rVSV) that expresses an individual filovirus glycoprotein (GP) in place of the VSV glycoprotein (G). The main concern with all replication-competent vaccines, including the rVSV filovirus GP vectors, is their safety. To address this concern, we performed a neurovirulence study using 21 cynomolgus macaques where the vaccines were administered intrathalamically. Seven animals received a rVSV vector expressing the Zaire ebolavirus (ZEBOV) GP; seven animals received a rVSV vector expressing the Lake Victoria marburgvirus (MARV) GP; three animals received rVSV-wild type (wt) vector, and four animals received vehicle control. Two of three animals given rVSV-wt showed severe neurological symptoms whereas animals receiving vehicle control, rVSV-ZEBOV-GP, or rVSV-MARV-GP did not develop these symptoms. Histological analysis revealed major lesions in neural tissues of all three rVSV-wt animals; however, no significant lesions were observed in any animals from the filovirus vaccine or vehicle control groups. These data strongly suggest that rVSV filovirus GP vaccine vectors lack the neurovirulence properties associated with the rVSV-wt parent vector and support their further development as a vaccine platform for human use.
- Published
- 2012
- Full Text
- View/download PDF
44. A small nonhuman primate model for filovirus-induced disease.
- Author
-
Carrion R Jr, Ro Y, Hoosien K, Ticer A, Brasky K, de la Garza M, Mansfield K, and Patterson JL
- Subjects
- Animals, Disease Models, Animal, Hemorrhagic Fever, Ebola mortality, Kidney pathology, Kidney virology, Liver pathology, Liver virology, Lung pathology, Lung virology, Marburg Virus Disease mortality, Monkey Diseases virology, Spleen pathology, Spleen virology, Viral Load, Callithrix virology, Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola pathology, Marburg Virus Disease pathology, Marburgvirus pathogenicity
- Abstract
Ebolavirus and Marburgvirus are members of the filovirus family and induce a fatal hemorrhagic disease in humans and nonhuman primates with 90% case fatality. To develop a small nonhuman primate model for filovirus disease, common marmosets (Callithrix jacchus) were intramuscularly inoculated with wild type Marburgvirus Musoke or Ebolavirus Zaire. The infection resulted in a systemic fatal disease with clinical and morphological features closely resembling human infection. Animals experienced weight loss, fever, high virus titers in tissue, thrombocytopenia, neutrophilia, high liver transaminases and phosphatases and disseminated intravascular coagulation. Evidence of a severe disseminated viral infection characterized principally by multifocal to coalescing hepatic necrosis was seen in EBOV animals. MARV-infected animals displayed only moderate fibrin deposition in the spleen. Lymphoid necrosis and lymphocytic depletion observed in spleen. These findings provide support for the use of the common marmoset as a small nonhuman primate model for filovirus induced hemorrhagic fever., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
45. Filovirus infection of STAT-1 knockout mice.
- Author
-
Raymond J, Bradfute S, and Bray M
- Subjects
- Animals, Ebola Vaccines administration & dosage, Ebola Vaccines immunology, Ebolavirus classification, Ebolavirus immunology, Female, Hemorrhagic Fever, Ebola prevention & control, Immunization Schedule, Immunoglobulin M blood, Male, Marburgvirus immunology, Mice, Mice, Knockout, Time Factors, Virulence, Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola immunology, Marburg Virus Disease immunology, Marburgvirus pathogenicity, STAT1 Transcription Factor genetics, STAT1 Transcription Factor metabolism
- Abstract
We evaluated the susceptibility to Ebola and Marburg virus infection of mice that cannot respond to interferon (IFN)-α/β and IFN-γ because of deletion of the STAT-1 gene. A mouse-adapted Zaire ebolavirus (ZEBOV) caused rapidly lethal disease; wild-type ZEBOV and Sudan Ebolavirus and 4 different Marburg virus strains produced severe, but more slowly progressive illness; and Reston Ebolavirus caused mild disease that was late in onset. The virulence of each agent was mirrored by the pace and severity of pathologic changes in the liver and lymphoid tissues. A virus-like particle vaccine elicited strong antibody responses but did not protect against mouse-adapted ZEBOV challenge.
- Published
- 2011
- Full Text
- View/download PDF
46. Antibody-dependent enhancement of Marburg virus infection.
- Author
-
Nakayama E, Tomabechi D, Matsuno K, Kishida N, Yoshida R, Feldmann H, and Takada A
- Subjects
- Animals, Antibodies, Monoclonal, Complement C1q metabolism, Epitopes immunology, Female, Gene Deletion, Glycoproteins chemistry, Glycoproteins genetics, Humans, Immune Sera, K562 Cells, Marburg Virus Disease virology, Marburgvirus pathogenicity, Mice, Mice, Inbred BALB C, Viral Proteins chemistry, Viral Proteins genetics, Viral Proteins immunology, Antibody-Dependent Enhancement, Glycoproteins immunology, Marburg Virus Disease immunology, Marburgvirus immunology
- Abstract
Background: Marburg virus (MARV) and Ebola virus (EBOV) cause severe hemorrhagic fever in primates. Earlier studies demonstrated that antibodies to particular epitopes on the glycoprotein (GP) of EBOV enhanced virus infectivity in vitro., Methods: To investigate this antibody-dependent enhancement (ADE) in MARV infection, we produced mouse antisera and monoclonal antibodies (mAbs) to the GPs of MARV strains Angola and Musoke., Results: The infectivity of vesicular stomatitis virus pseudotyped with Angola GP in K562 cells was significantly enhanced in the presence of Angola GP antisera, whereas only minimal ADE activity was seen with Musoke GP antisera. This difference correlated with the percentage of hybridoma clones producing infectivity-enhancing mAbs. Using mAbs to MARV GP, we identified 3 distinct ADE epitopes in the mucinlike region on Angola GP. Interestingly, some of these antibodies bound to both Angola and Musoke GPs but showed significantly higher ADE activity for strain Angola. ADE activity depended on epitopes in the mucinlike region and glycine at amino acid position 547, present in the Angola but absent in the Musoke GP., Conclusions: These results suggest a possible link between ADE and MARV pathogenicity and provide new insights into the mechanisms underlying ADE entry of filoviruses.
- Published
- 2011
- Full Text
- View/download PDF
47. Current perspectives on the phylogeny of Filoviridae.
- Author
-
Barrette RW, Xu L, Rowland JM, and McIntosh MT
- Subjects
- Animals, Disease Reservoirs virology, Ebolavirus classification, Ebolavirus genetics, Ebolavirus pathogenicity, Filoviridae pathogenicity, Filoviridae Infections transmission, Filoviridae Infections virology, Genome, Viral, Humans, Marburgvirus classification, Marburgvirus genetics, Marburgvirus pathogenicity, Phylogeny, Swine virology, Filoviridae classification, Filoviridae genetics
- Abstract
Sporadic fatal outbreaks of disease in humans and non-human primates caused by Ebola or Marburg viruses have driven research into the characterization of these viruses with the hopes of identifying host tropisms and potential reservoirs. Such an understanding of the relatedness of newly discovered filoviruses may help to predict risk factors for outbreaks of hemorrhagic disease in humans and/or non-human primates. Recent discoveries such as three distinct genotypes of Reston ebolavirus, unexpectedly discovered in domestic swine in the Philippines; as well as a new species, Bundibugyo ebolavirus; the recent discovery of Lloviu virus as a potential new genus, Cuevavirus, within Filoviridae; and germline integrations of filovirus-like sequences in some animal species bring new insights into the relatedness of filoviruses, their prevalence and potential for transmission to humans. These new findings reveal that filoviruses are more diverse and may have had a greater influence on the evolution of animals than previously thought. Herein we review these findings with regard to the implications for understanding the host range, prevalence and transmission of Filoviridae., (Published by Elsevier B.V.)
- Published
- 2011
- Full Text
- View/download PDF
48. Filoviral immune evasion mechanisms.
- Author
-
Ramanan P, Shabman RS, Brown CS, Amarasinghe GK, Basler CF, and Leung DW
- Subjects
- Adaptive Immunity, Animals, Ebolavirus metabolism, Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola virology, Humans, Immunity, Innate, Interferon Type I metabolism, Marburg Virus Disease virology, Marburgvirus metabolism, Marburgvirus pathogenicity, Phosphorylation, Signal Transduction, Virus Replication, Ebolavirus immunology, Hemorrhagic Fever, Ebola immunology, Immune Evasion immunology, Marburg Virus Disease immunology, Marburgvirus immunology, Viral Proteins metabolism
- Abstract
The Filoviridae family of viruses, which includes the genera Ebolavirus (EBOV) and Marburgvirus (MARV), causes severe and often times lethal hemorrhagic fever in humans. Filoviral infections are associated with ineffective innate antiviral responses as a result of virally encoded immune antagonists, which render the host incapable of mounting effective innate or adaptive immune responses. The Type I interferon (IFN) response is critical for establishing an antiviral state in the host cell and subsequent activation of the adaptive immune responses. Several filoviral encoded components target Type I IFN responses, and this innate immune suppression is important for viral replication and pathogenesis. For example, EBOV VP35 inhibits the phosphorylation of IRF-3/7 by the TBK-1/IKKε kinases in addition to sequestering viral RNA from detection by RIG-I like receptors. MARV VP40 inhibits STAT1/2 phosphorylation by inhibiting the JAK family kinases. EBOV VP24 inhibits nuclear translocation of activated STAT1 by karyopherin-α. The examples also represent distinct mechanisms utilized by filoviral proteins in order to counter immune responses, which results in limited IFN-α/β production and downstream signaling.
- Published
- 2011
- Full Text
- View/download PDF
49. The cytoplasmic domain of Marburg virus GP modulates early steps of viral infection.
- Author
-
Mittler E, Kolesnikova L, Hartlieb B, Davey R, and Becker S
- Subjects
- Animals, Antibodies, Monoclonal, Cytoplasm, Fluorescent Antibody Technique, Indirect, Glycosylation, HEK293 Cells, Humans, Luciferases, Marburgvirus metabolism, Marburgvirus pathogenicity, Mutation, Protein Structure, Tertiary, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Viral Fusion Proteins chemistry, Viral Fusion Proteins genetics, Viral Fusion Proteins immunology, Viral Fusion Proteins metabolism, Viral Matrix Proteins metabolism, Marburgvirus physiology, Viral Envelope Proteins chemistry, Viral Envelope Proteins metabolism, Virus Internalization
- Abstract
Marburg virus infection is mediated by the only viral surface protein, GP, a trimeric type I transmembrane protein. While its ectodomain mediates receptor binding and fusion of viral and cellular membranes and its transmembrane domain is essential for the recruitment of GP into budding particles by the matrix protein VP40, the role of the short cytoplasmic domain has remained enigmatic. Here we show that a missing cytoplasmic domain did not impair trimerization, intracellular transport, or incorporation of GP into infectious Marburg virus-like particles (iVLPs) but altered the glycosylation pattern as well as the recognition of GP by neutralizing antibodies. These results suggest that subtle conformational changes took place in the ectodomain. To investigate the function of the cytoplasmic domain during viral entry, a novel entry assay was established to monitor the uptake of filamentous VLPs by measuring the occurrence of luciferase-labeled viral nucleocapsids in the cytosol of target cells. This quantitative assay showed that the entry process of VLPs incorporating GP missing its cytoplasmic domain (GPΔCD) was impaired. Supporting these results, iVLPs incorporating a mutant GP missing its cytoplasmic domain were significantly less infectious than iVLPs containing wild-type GP. Taken together, the data indicate that the absence of the short cytoplasmic domain of Marburg virus GP may induce conformational changes in the ectodomain which impact the filoviral entry process.
- Published
- 2011
- Full Text
- View/download PDF
50. Ebola and Marburg haemorrhagic fever viruses: major scientific advances, but a relatively minor public health threat for Africa.
- Author
-
Leroy EM, Gonzalez JP, and Baize S
- Subjects
- Africa epidemiology, Animals, Communicable Disease Control methods, Hemorrhagic Fever, Ebola mortality, Hemorrhagic Fever, Ebola virology, Humans, Incidence, Marburg Virus Disease mortality, Marburg Virus Disease virology, Disease Outbreaks, Ebolavirus pathogenicity, Hemorrhagic Fever, Ebola epidemiology, Hemorrhagic Fever, Ebola veterinary, Marburg Virus Disease epidemiology, Marburgvirus pathogenicity
- Abstract
Ebola and Marburg viruses are the only members of the Filoviridae family (order Mononegavirales), a group of viruses characterized by a linear, non-segmented, single-strand negative RNA genome. They are among the most virulent pathogens for humans and great apes, causing acute haemorrhagic fever and death within a matter of days. Since their discovery 50 years ago, filoviruses have caused only a few outbreaks, with 2317 clinical cases and 1671 confirmed deaths, which is negligible compared with the devastation caused by malnutrition and other infectious diseases prevalent in Africa (malaria, cholera, AIDS, dengue, tuberculosis …). Yet considerable human and financial resourses have been devoted to research on these viruses during the past two decades, partly because of their potential use as bioweapons. As a result, our understanding of the ecology, host interactions, and control of these viruses has improved considerably., (© 2011 The Authors. Clinical Microbiology and Infection © 2011 European Society of Clinical Microbiology and Infectious Diseases.)
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.