Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Microbiol Spectr ; 11(3): e0349422, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37036346

ABSTRACT

Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. The pathogenesis of MVD remains poorly understood, partially due to the low number of cases that can be studied, the absence of state-of-the-art medical equipment in areas where cases are reported, and limitations on the number of animals that can be safely used in experimental studies under maximum containment animal biosafety level 4 conditions. Medical imaging modalities, such as whole-body computed tomography (CT), may help to describe disease progression in vivo, potentially replacing ethically contentious and logistically challenging serial euthanasia studies. Towards this vision, we performed a pilot study, during which we acquired whole-body CT images of 6 rhesus monkeys before and 7 to 9 days after intramuscular MARV exposure. We identified imaging abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to clinical, virological, and gross pathological hallmarks of MVD in this animal model. Quantitative image analysis indicated hepatomegaly with a significant reduction in organ density (indicating fatty infiltration of the liver), splenomegaly, and edema that corresponded with gross pathological and histopathological findings. Our results indicated that CT imaging could be used to verify and quantify typical MVD pathogenesis versus altered, diminished, or absent disease severity or progression in the presence of candidate medical countermeasures, thus possibly reducing the number of animals needed and eliminating serial euthanasia. IMPORTANCE Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. Much is unknown about disease progression and, thus, prevention and treatment options are limited. Medical imaging modalities, such as whole-body computed tomography (CT), have the potential to improve understanding of MVD pathogenesis. Our study used CT to identify abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to known clinical signs of MVD in this animal model. Our results indicated that CT imaging and analyses could be used to elucidate pathogenesis and possibly assess the efficacy of candidate treatments.


Subject(s)
Marburg Virus Disease , Marburgvirus , Humans , Animals , Marburg Virus Disease/diagnostic imaging , Marburg Virus Disease/pathology , Pilot Projects , Tomography, X-Ray Computed , Disease Progression , Primates
2.
J Infect Dis ; 222(11): 1894-1901, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32479636

ABSTRACT

Marburg virus (MARV) is a filovirus with documented human case-fatality rates of up to 90%. Here, we evaluated the therapeutic efficacy of remdesivir (GS-5734) in nonhuman primates experimentally infected with MARV. Beginning 4 or 5 days post inoculation, cynomolgus macaques were treated once daily for 12 days with vehicle, 5 mg/kg remdesivir, or a 10-mg/kg loading dose followed by 5 mg/kg remdesivir. All vehicle-control animals died, whereas 83% of animals receiving a 10-mg/kg loading dose of remdesivir survived, as did 50% of animals receiving a 5-mg/kg remdesivir regimen. Remdesivir-treated animals exhibited improved clinical scores, lower plasma viral RNA, and improved markers of kidney function, liver function, and coagulopathy versus vehicle-control animals. The small molecule remdesivir showed therapeutic efficacy in this Marburg virus disease model with treatment initiation 5 days post inoculation, supporting further assessment of remdesivir for the treatment of Marburg virus disease in humans.


Subject(s)
Antimetabolites/therapeutic use , Antiviral Agents/therapeutic use , Marburg Virus Disease/drug therapy , Marburgvirus/drug effects , Monkey Diseases/drug therapy , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Animals , Disease Models, Animal , Female , Kaplan-Meier Estimate , Macaca fascicularis , Male , Marburg Virus Disease/mortality , Marburg Virus Disease/pathology , Marburg Virus Disease/virology , Monkey Diseases/mortality , Monkey Diseases/pathology , Monkey Diseases/virology , RNA, Viral
3.
Nat Rev Microbiol ; 18(7): 365-378, 2020 07.
Article in English | MEDLINE | ID: mdl-32367066

ABSTRACT

Filoviruses such as Ebola virus continue to pose a substantial health risk to humans. Advances in the sequencing and functional characterization of both pathogen and host genomes have provided a wealth of knowledge to clinicians, epidemiologists and public health responders during outbreaks of high-consequence viral disease. Here, we describe how genomics has been historically used to investigate Ebola virus disease outbreaks and how new technologies allow for rapid, large-scale data generation at the point of care. We highlight how genomics extends beyond consensus-level sequencing of the virus to include intra-host viral transcriptomics and the characterization of host responses in acute and persistently infected patients. Similar genomics techniques can also be applied to the characterization of non-human primate animal models and to known natural reservoirs of filoviruses, and metagenomic sequencing can be the key to the discovery of novel filoviruses. Finally, we outline the importance of reverse genetics systems that can swiftly characterize filoviruses as soon as their genome sequences are available.


Subject(s)
Ebolavirus/genetics , Hemorrhagic Fever, Ebola/epidemiology , Host-Pathogen Interactions/genetics , Marburg Virus Disease/epidemiology , Marburgvirus/genetics , Africa/epidemiology , Animals , Disease Outbreaks , Female , Genome, Viral/genetics , Genomics/methods , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/transmission , Humans , Male , Marburg Virus Disease/pathology , Marburg Virus Disease/transmission , Molecular Epidemiology/methods , Reverse Genetics/methods , Virus Replication/genetics
4.
PLoS Negl Trop Dis ; 13(3): e0007257, 2019 03.
Article in English | MEDLINE | ID: mdl-30883555

ABSTRACT

INTRODUCTION: In October 2017, a blood sample from a resident of Kween District, Eastern Uganda, tested positive for Marburg virus. Within 24 hour of confirmation, a rapid outbreak response was initiated. Here, we present results of epidemiological and laboratory investigations. METHODS: A district task force was activated consisting of specialised teams to conduct case finding, case management and isolation, contact listing and follow up, sample collection and testing, and community engagement. An ecological investigation was also carried out to identify the potential source of infection. Virus isolation and Next Generation sequencing were performed to identify the strain of Marburg virus. RESULTS: Seventy individuals (34 MVD suspected cases and 36 close contacts of confirmed cases) were epidemiologically investigated, with blood samples tested for MVD. Only four cases met the MVD case definition; one was categorized as a probable case while the other three were confirmed cases. A total of 299 contacts were identified; during follow- up, two were confirmed as MVD. Of the four confirmed and probable MVD cases, three died, yielding a case fatality rate of 75%. All four cases belonged to a single family and 50% (2/4) of the MVD cases were female. All confirmed cases had clinical symptoms of fever, vomiting, abdominal pain and bleeding from body orifices. Viral sequences indicated that the Marburg virus strain responsible for this outbreak was closely related to virus strains previously shown to be circulating in Uganda. CONCLUSION: This outbreak of MVD occurred as a family cluster with no additional transmission outside of the four related cases. Rapid case detection, prompt laboratory testing at the Uganda National VHF Reference Laboratory and presence of pre-trained, well-prepared national and district rapid response teams facilitated the containment and control of this outbreak within one month, preventing nationwide and global transmission of the disease.


Subject(s)
Clinical Laboratory Techniques/methods , Communicable Disease Control/methods , Disease Outbreaks , Marburg Virus Disease/epidemiology , Marburg Virus Disease/pathology , Marburgvirus/isolation & purification , Adult , Animals , Cluster Analysis , Disease Transmission, Infectious/prevention & control , Family Health , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Marburg Virus Disease/mortality , Middle Aged , Mortality , Uganda/epidemiology , Virus Cultivation
5.
Viruses ; 11(3)2019 03 02.
Article in English | MEDLINE | ID: mdl-30832364

ABSTRACT

Egyptian rousette bats (Rousettus aegyptiacus) are natural reservoir hosts of Marburg virus (MARV), and Ravn virus (RAVV; collectively called marburgviruses) and have been linked to human cases of Marburg virus disease (MVD). We investigated the clinical and pathologic effects of experimental MARV infection in Egyptian rousettes through a serial euthanasia study and found clear evidence of mild but transient disease. Three groups of nine, captive-born, juvenile male bats were inoculated subcutaneously with 10,000 TCID50 of Marburg virus strain Uganda 371Bat2007, a minimally passaged virus originally isolated from a wild Egyptian rousette. Control bats (n = 3) were mock-inoculated. Three animals per day were euthanized at 3, 5⁻10, 12 and 28 days post-inoculation (DPI); controls were euthanized at 28 DPI. Blood chemistry analyses showed a mild, statistically significant elevation in alanine aminotransferase (ALT) at 3, 6 and 7 DPI. Lymphocyte and monocyte counts were mildly elevated in inoculated bats after 9 DPI. Liver histology revealed small foci of inflammatory infiltrate in infected bats, similar to lesions previously described in wild, naturally-infected bats. Liver lesion severity scores peaked at 7 DPI, and were correlated with both ALT and hepatic viral RNA levels. Immunohistochemical staining detected infrequent viral antigen in liver (3⁻8 DPI, n = 8), spleen (3⁻7 DPI, n = 8), skin (inoculation site; 3⁻12 DPI, n = 20), lymph nodes (3⁻10 DPI, n = 6), and oral submucosa (8⁻9 DPI, n = 2). Viral antigen was present in histiocytes, hepatocytes and mesenchymal cells, and in the liver, antigen staining co-localized with inflammatory foci. These results show the first clear evidence of very mild disease caused by a filovirus in a reservoir bat host and provide support for our experimental model of this virus-reservoir host system.


Subject(s)
Chiroptera/virology , Disease Reservoirs/virology , Liver/virology , Marburg Virus Disease/immunology , Alanine Transaminase/blood , Animals , Antibodies, Viral/blood , Immunoglobulin G/blood , Immunohistochemistry , Liver/pathology , Lymphocyte Count , Male , Marburg Virus Disease/pathology , Marburgvirus , RNA, Viral/genetics , Subcutaneous Absorption
6.
Lab Med ; 50(1): 16-28, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30085179

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.


Subject(s)
Marburg Virus Disease/epidemiology , Marburgvirus/pathogenicity , Animals , Humans , Marburg Virus Disease/pathology , Marburg Virus Disease/therapy , Marburg Virus Disease/transmission , Marburgvirus/genetics
7.
Clin Microbiol Infect ; 21S: e28-e31, 2019 Apr.
Article in English | MEDLINE | ID: mdl-24816494

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.


Subject(s)
Marburg Virus Disease/diagnosis , Marburg Virus Disease/prevention & control , Marburgvirus/isolation & purification , Travel-Related Illness , Animals , Disease Outbreaks/prevention & control , Early Diagnosis , Humans , Infection Control , Marburg Virus Disease/pathology , Marburg Virus Disease/therapy , Marburgvirus/pathogenicity
8.
Viruses ; 10(11)2018 11 21.
Article in English | MEDLINE | ID: mdl-30469360

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.


Subject(s)
Macaca , Marburg Virus Disease/pathology , Marburgvirus/pathogenicity , Animals , Disease Models, Animal , Injections, Intramuscular , Retrospective Studies , Survival Analysis , United States , Virulence
9.
J Infect Dis ; 218(suppl_5): S438-S447, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30192975

ABSTRACT

Marburg virus (MARV; family Filoviridae) causes sporadic outbreaks of Marburg hemorrhagic fever in sub-Saharan Africa with case fatality rates reaching 90%. Wild-type filoviruses, including MARV and the closely related Ebola virus, are unable to suppress the type I interferon response in rodents, and therefore require adaptation of the viruses to cause disease in immunocompetent animals. In the current study, we demonstrate that STAT2 knockout Syrian hamsters are susceptible to infection with different wild-type MARV variants. MARV Musoke causes a robust and systemic infection resulting in lethal disease. Histopathological findings share features similar to those observed in human patients and other animal models of filovirus infection. Reverse-transcription polymerase chain reaction analysis of host transcripts shows a dysregulation of the innate immune response. Our results demonstrate that the STAT2 knockout hamster represents a novel small animal model of severe MARV infection and disease without the requirement for virus adaptation.


Subject(s)
Marburg Virus Disease/etiology , STAT2 Transcription Factor/physiology , Animals , Cricetinae , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility , Female , Male , Marburg Virus Disease/immunology , Marburg Virus Disease/pathology
10.
J Infect Dis ; 218(suppl_5): S458-S465, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30215737

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.


Subject(s)
Marburg Virus Disease/etiology , Marburgvirus/pathogenicity , Animals , Apoptosis , Hepatocytes/pathology , Macaca fascicularis , Macaca mulatta , Macrophages/pathology , Male , Marburg Virus Disease/immunology , Marburg Virus Disease/pathology , Phenotype
11.
J Infect Dis ; 218(suppl_5): S423-S433, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30053050

ABSTRACT

Previously, several studies have been performed to delineate the development and progression of Marburg virus infection in nonhuman primates (NHPs), primarily to clarify the mechanisms of severe (fatal) disease. After the 2013-2016 Ebola virus disease (EVD) epidemic in Western Africa, there has been a reassessment of the available filovirus animal models and the utility of these to faithfully recapitulate human disease. The high lethality of the NHP models has raised doubts as to their ability to provide meaningful data for the full spectrum of disease observed in humans. Of particular interest are the etiologic and pathophysiologic mechanisms underlying postconvalescent sequelae observed in human survivors of EVD and Marburg virus disease (MVD). In the current study, we evaluated the lesions of MVD in NHPs; however, in contrast to previous studies, we focused on the potential for development of sequelae similar to those reported in human survivors of MVD and EVD. We found that during acute MVD in the macaque model, there is frequent inflammation of peripheral nerves, autonomic ganglia, and the iris of the eye. Furthermore, we demonstrate viral infection of the ocular ciliary body and retina, testis, epididymis, ovary, oviduct, uterine endometrium, prostate, and mammary gland. These findings are relevant for both development of postconvalescent sequelae and the natural transmission of virus.


Subject(s)
Marburg Virus Disease/pathology , Animals , Disease Models, Animal , Eye/pathology , Female , Ganglia/pathology , Humans , Macaca mulatta , Male , Mammary Glands, Human/pathology , Peripheral Nerves/pathology , Urogenital System/pathology
12.
J Infect Dis ; 218(suppl_5): S448-S452, 2018 11 22.
Article in English | MEDLINE | ID: mdl-29955887

ABSTRACT

The domestic ferret was recently described as a uniformly lethal model for 3 species of Ebolavirus. More importantly, this new model utilizes nonadapted wild-type Ebolaviruses. Here, in a proof-of-concept study, we infected ferrets with different variants of the closely related Marburg and Ravn viruses using different doses and routes of exposure. Although ferrets produced a neutralizing humoral response to challenge, we did not observe disease or viremia in any animal. The lack of disease in ferrets underscores the notion that differential mechanisms to immunity among filoviruses exist and may provide a model to better understand how differences contribute to disease.


Subject(s)
Disease Models, Animal , Marburg Virus Disease/immunology , Animals , Antibodies, Viral/blood , Female , Ferrets , Marburg Virus Disease/pathology , Marburgvirus/immunology
13.
Cell ; 173(5): 1098-1110.e18, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29706541

ABSTRACT

Bats harbor many viruses asymptomatically, including several notorious for causing extreme virulence in humans. To identify differences between antiviral mechanisms in humans and bats, we sequenced, assembled, and analyzed the genome of Rousettus aegyptiacus, a natural reservoir of Marburg virus and the only known reservoir for any filovirus. We found an expanded and diversified KLRC/KLRD family of natural killer cell receptors, MHC class I genes, and type I interferons, which dramatically differ from their functional counterparts in other mammals. Such concerted evolution of key components of bat immunity is strongly suggestive of novel modes of antiviral defense. An evaluation of the theoretical function of these genes suggests that an inhibitory immune state may exist in bats. Based on our findings, we hypothesize that tolerance of viral infection, rather than enhanced potency of antiviral defenses, may be a key mechanism by which bats asymptomatically host viruses that are pathogenic in humans.


Subject(s)
Chiroptera/genetics , Genome , Immunity, Innate/genetics , Amino Acid Sequence , Animals , Cell Line , Chiroptera/classification , Chiroptera/immunology , Chromosome Mapping , Disease Reservoirs/virology , Egypt , Evolution, Molecular , Genetic Variation , Histocompatibility Antigens Class I/classification , Histocompatibility Antigens Class I/genetics , Humans , Interferon Type I/classification , Interferon Type I/genetics , Marburg Virus Disease/immunology , Marburg Virus Disease/pathology , Marburgvirus/physiology , NK Cell Lectin-Like Receptor Subfamily C/chemistry , NK Cell Lectin-Like Receptor Subfamily C/classification , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily D/chemistry , NK Cell Lectin-Like Receptor Subfamily D/classification , NK Cell Lectin-Like Receptor Subfamily D/genetics , Phylogeny , Sequence Alignment
14.
Zool Res ; 39(1): 32-41, 2018 Jan 18.
Article in English | MEDLINE | ID: mdl-29511143

ABSTRACT

The Angolan strain of Marburg virus (MARV/Ang) can cause lethal disease in humans with a case fatality rate of up to 90%, but infection of immunocompetent rodents do not result in any observable symptoms. Our previous work includes the development and characterization of a MARV/Ang variant that can cause lethal disease in mice (MARV/Ang-MA), with the aim of using this tool to screen for promising prophylactic and therapeutic candidates. An intermediate animal model is needed to confirm any findings from mice studies before testing in the gold-standard non-human primate (NHP) model. In this study, we serially passaged the clinical isolate of MARV/Ang in the livers and spleens of guinea pigs until a variant emerged that causes 100% lethality in guinea pigs (MARV/Ang-GA). Animals infected with MARV/Ang-GA showed signs of filovirus infection including lymphocytopenia, thrombocytopenia, and high viremia leading to spread to major organs, including the liver, spleen, lungs, and kidneys. The MARV/Ang-GA guinea pigs died between 7-9 days after infection, and the LD50 was calculated to be 1.1×10-1 TCID50 (median tissue culture infective dose). Mutations in MARV/Ang-GA were identified and compared to sequences of known rodent-adapted MARV/Ang variants, which may benefit future studies characterizing important host adaptation sites in the MARV/Ang viral genome.


Subject(s)
Marburg Virus Disease/etiology , Marburgvirus , Animals , Disease Models, Animal , Female , Guinea Pigs/virology , Marburg Virus Disease/pathology , Marburg Virus Disease/virology , Reverse Transcriptase Polymerase Chain Reaction , Viral Load , Viremia/pathology , Viremia/virology
15.
Antiviral Res ; 151: 97-104, 2018 03.
Article in English | MEDLINE | ID: mdl-29289666

ABSTRACT

Favipiravir is a broad-spectrum antiviral agent that has demonstrated efficacy against Ebola virus (EBOV) in rodents. However, there are no published reports of favipiravir efficacy for filovirus infection of nonhuman primates (NHPs). Here we evaluated the pharmacokinetic profile of favipiravir in NHPs, as well as in vivo efficacy against two filoviruses, EBOV and Marburg virus (MARV). While no survival benefit was observed in two studies employing once- or twice-daily oral dosing of favipiravir during EBOV infection of NHPs, an antiviral effect was observed in terms of extended time-to-death and reduced levels of viral RNA. However, oral dosing in biosafety level-4 (BSL-4) presents logistical and technical challenges, and repeated anesthesia events may potentially worsen survival outcome in animals. For the third study of treatment of MARV infection, we therefore made use of catheters, jackets, and tethers for intravenous (IV) dosing and blood collection, which minimized the requirement for repeated anesthesia events. When MARV infection was treated with IV favipiravir, five of six animals (83%) survived infection, while all untreated NHPs succumbed. An accompanying report presents the results of favipiravir treatment of EBOV infection in mice.


Subject(s)
Amides/administration & dosage , Amides/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Marburg Virus Disease/drug therapy , Marburgvirus/drug effects , Pyrazines/administration & dosage , Pyrazines/pharmacology , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Male , Marburg Virus Disease/pathology , Marburg Virus Disease/virology , Primates , RNA, Viral/blood , Survival Analysis , Viral Load/drug effects
16.
J Clin Invest ; 127(12): 4437-4448, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29106386

ABSTRACT

Ebolaviruses and marburgviruses belong to the family Filoviridae and cause high lethality in infected patients. There are currently no licensed filovirus vaccines or antiviral therapies. The development of broad-spectrum therapies against members of the Marburgvirus genus, including Marburg virus (MARV) and Ravn virus (RAVV), is difficult because of substantial sequence variability. RNAi therapeutics offer a potential solution, as identification of conserved target nucleotide sequences may confer activity across marburgvirus variants. Here, we assessed the therapeutic efficacy of lipid nanoparticle (LNP) delivery of a single nucleoprotein-targeting (NP-targeting) siRNA in nonhuman primates at advanced stages of MARV or RAVV disease to mimic cases in which patients begin treatment for fulminant disease. Sixteen rhesus monkeys were lethally infected with MARV or RAVV and treated with NP siRNA-LNP, with MARV-infected animals beginning treatment four or five days after infection and RAVV-infected animals starting treatment three or six days after infection. While all untreated animals succumbed to disease, NP siRNA-LNP treatment conferred 100% survival of RAVV-infected macaques, even when treatment began just 1 day prior to the death of the control animals. In MARV-infected animals, day-4 treatment initiation resulted in 100% survival, and day-5 treatment resulted in 50% survival. These results identify a single siRNA therapeutic that provides broad-spectrum protection against both MARV and RAVV.


Subject(s)
Drug Delivery Systems/methods , Marburg Virus Disease/drug therapy , Marburgvirus , Nanoparticles/therapeutic use , RNA, Small Interfering/pharmacology , Animals , Macaca mulatta , Marburg Virus Disease/metabolism , Marburg Virus Disease/pathology , Nanoparticles/chemistry , RNA, Small Interfering/chemistry
17.
Sci Rep ; 7(1): 3390, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28611428

ABSTRACT

Marburg virus (MARV) has caused outbreaks of filoviral hemorrhagic fever since its discovery in 1967. The largest and deadliest outbreak occurred in Angola in 2005, with 252 cases and 227 deaths. In 2014, we developed a mouse-adapted MARV, Angola variant through serial passaging in mice. The mouse-adapted MARV exhibits many of the hallmarks of MARV disease in humans. By applying deep-sequencing to every passage of the virus, we are able to study virus evolution in this host with surprising precision. We show that two regions go through substantial changes: the intergenic region between NP and VP35, as well as the first 100 amino acids of the VP40 protein. Our results also reveal that there were profound changes during the production of the final virus stock in cell culture. Overall, our results show that a handful of regions carry most of the mutations acquired during the adaptation of the virus to a new host and that many mutations become fixed very early during the adaptation process.


Subject(s)
Adaptation, Biological/genetics , Genome, Viral , High-Throughput Nucleotide Sequencing/methods , Marburg Virus Disease/pathology , Marburg Virus Disease/virology , Marburgvirus/genetics , Viral Proteins/genetics , Animals , Cells, Cultured , Marburg Virus Disease/genetics , Marburgvirus/growth & development , Marburgvirus/isolation & purification , Mice , RNA, Viral/genetics , Serial Passage , Viral Load
18.
PLoS Pathog ; 13(1): e1006132, 2017 01.
Article in English | MEDLINE | ID: mdl-28076420

ABSTRACT

Ebola (EBOV) and Marburg (MARV) viruses are members of the Filoviridae family which cause outbreaks of hemorrhagic fever. The filovirus VP40 matrix protein is essential for virus assembly and budding, and its PPxY L-domain motif interacts with WW-domains of specific host proteins, such as Nedd4 and ITCH, to facilitate the late stage of virus-cell separation. To identify additional WW-domain-bearing host proteins that interact with VP40, we used an EBOV PPxY-containing peptide to screen an array of 115 mammalian WW-domain-bearing proteins. Using this unbiased approach, we identified BCL2 Associated Athanogene 3 (BAG3), a member of the BAG family of molecular chaperone proteins, as a specific VP40 PPxY interactor. Here, we demonstrate that the WW-domain of BAG3 interacts with the PPxY motif of both EBOV and MARV VP40 and, unexpectedly, inhibits budding of both eVP40 and mVP40 virus-like particles (VLPs), as well as infectious VSV-EBOV recombinants. BAG3 is a stress induced protein that regulates cellular protein homeostasis and cell survival through chaperone-mediated autophagy (CMA). Interestingly, our results show that BAG3 alters the intracellular localization of VP40 by sequestering VP40 away from the plasma membrane. As BAG3 is the first WW-domain interactor identified that negatively regulates budding of VP40 VLPs and infectious virus, we propose that the chaperone-mediated autophagy function of BAG3 represents a specific host defense strategy to counteract the function of VP40 in promoting efficient egress and spread of virus particles.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Autophagy/physiology , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/transmission , Marburg Virus Disease/transmission , Marburgvirus/metabolism , Viral Matrix Proteins/metabolism , Virus Release/genetics , Animals , Autophagy/genetics , Cell Line, Tumor , Cell Survival/genetics , Cricetinae , Ebolavirus/genetics , Endosomal Sorting Complexes Required for Transport , HEK293 Cells , HeLa Cells , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Humans , Marburg Virus Disease/pathology , Marburg Virus Disease/virology , Marburgvirus/genetics , Nedd4 Ubiquitin Protein Ligases , Proline/analogs & derivatives , Proline/metabolism , Protein Structure, Tertiary , Protein Transport/physiology , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
19.
Sci Rep ; 6: 39214, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27976688

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.


Subject(s)
Marburg Virus Disease/pathology , Marburgvirus/pathogenicity , 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
20.
Viruses ; 7(7): 3420-42, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26120867

ABSTRACT

The Egyptian rousette bat (Rousettus aegyptiacus) is a natural reservoir for marburgviruses and a consistent source of virus spillover to humans. Cumulative evidence suggests various bat species may also transmit ebolaviruses. We investigated the susceptibility of Egyptian rousettes to each of the five known ebolaviruses (Sudan, Ebola, Bundibugyo, Taï Forest, and Reston), and compared findings with Marburg virus. In a pilot study, groups of four juvenile bats were inoculated with one of the ebolaviruses or Marburg virus. In ebolavirus groups, viral RNA tissue distribution was limited, and no bat became viremic. Sudan viral RNA was slightly more widespread, spurring a second, 15-day Sudan virus serial euthanasia study. Low levels of Sudan viral RNA disseminated to multiple tissues at early time points, but there was no viremia or shedding. In contrast, Marburg virus RNA was widely disseminated, with viremia, oral and rectal shedding, and antigen in spleen and liver. This is the first experimental infection study comparing tissue tropism, viral shedding, and clinical and pathologic effects of six different filoviruses in the Egyptian rousette, a known marburgvirus reservoir. Our results suggest Egyptian rousettes are unlikely sources for ebolaviruses in nature, and support a possible single filovirus-single reservoir host relationship.


Subject(s)
Chiroptera/virology , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Marburg Virus Disease/virology , Marburgvirus/physiology , Animals , Disease Models, Animal , Disease Reservoirs/virology , Ebolavirus/classification , Hemorrhagic Fever, Ebola/pathology , Humans , Liver/pathology , Liver/virology , Marburg Virus Disease/pathology , Spleen/pathology , Spleen/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...