Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
BMC Biotechnol ; 24(1): 45, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970027

ABSTRACT

Marburg virus (MARV) is a highly contagious and virulent agent belonging to Filoviridae family. MARV causes severe hemorrhagic fever in humans and non-human primates. Owing to its highly virulent nature, preventive approaches are promising for its control. There is currently no approved drug or vaccine against MARV, and management mainly involves supportive care to treat symptoms and prevent complications. Our aim was to design a novel multi-epitope vaccine (MEV) against MARV using immunoinformatics studies. In this study, various proteins (VP35, VP40 and glycoprotein precursor) were used and potential epitopes were selected. CTL and HTL epitopes covered 79.44% and 70.55% of the global population, respectively. The designed MEV construct was stable and expressed in Escherichia coli (E. coli) host. The physicochemical properties were also acceptable. MARV MEV candidate could predict comprehensive immune responses such as those of humoral and cellular in silico. Additionally, efficient interaction to toll-like receptor 3 (TLR3) and its agonist (ß-defensin) was predicted. There is a need for validation of these results using further in vitro and in vivo studies.


Subject(s)
Computational Biology , Marburg Virus Disease , Marburgvirus , Viral Vaccines , Marburgvirus/immunology , Marburg Virus Disease/prevention & control , Marburg Virus Disease/immunology , Viral Vaccines/immunology , Computational Biology/methods , Animals , Humans , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes/immunology , Epitopes/genetics , Epitopes/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Immunoinformatics
2.
J Virol ; 98(7): e0015524, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38832790

ABSTRACT

Marburg virus infection in humans is associated with case fatality rates that can reach up to 90%, but to date, there are no approved vaccines or monoclonal antibody (mAb) countermeasures. Here, we immunized Rhesus macaques with multivalent combinations of filovirus glycoprotein (GP) antigens belonging to Marburg, Sudan, and Ebola viruses to generate monospecific and cross-reactive antibody responses against them. From the animal that developed the highest titers of Marburg virus GP-specific neutralizing antibodies, we sorted single memory B cells using a heterologous Ravn virus GP probe and cloned and characterized a panel of 34 mAbs belonging to 28 unique lineages. Antibody specificities were assessed by overlapping pepscan and binding competition analyses, revealing that roughly a third of the lineages mapped to the conserved receptor binding region, including potent neutralizing lineages that were confirmed by negative stain electron microscopy to target this region. Additional lineages targeted a protective region on GP2, while others were found to possess cross-filovirus reactivity. Our study advances the understanding of orthomarburgvirus glycoprotein antigenicity and furthers efforts to develop candidate antibody countermeasures against these lethal viruses. IMPORTANCE: Marburg viruses were the first filoviruses characterized to emerge in humans in 1967 and cause severe hemorrhagic fever with average case fatality rates of ~50%. Although mAb countermeasures have been approved for clinical use against the related Ebola viruses, there are currently no approved countermeasures against Marburg viruses. We successfully isolated a panel of orthomarburgvirus GP-specific mAbs from a macaque immunized with a multivalent combination of filovirus antigens. Our analyses revealed that roughly half of the antibodies in the panel mapped to regions on the glycoprotein shown to protect from infection, including the host cell receptor binding domain and a protective region on the membrane-anchoring subunit. Other antibodies in the panel exhibited broad filovirus GP recognition. Our study describes the discovery of a diverse panel of cross-reactive macaque antibodies targeting orthomarburgvirus and other filovirus GPs and provides candidate immunotherapeutics for further study and development.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Cross Reactions , Macaca mulatta , Marburg Virus Disease , Marburgvirus , Animals , Marburgvirus/immunology , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Antibodies, Monoclonal/immunology , Marburg Virus Disease/immunology , Marburg Virus Disease/prevention & control , Cross Reactions/immunology , Glycoproteins/immunology , Viral Envelope Proteins/immunology , Immunization , Humans , Ebolavirus/immunology , Antigens, Viral/immunology
3.
PLoS Pathog ; 20(6): e1012262, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924060

ABSTRACT

Viral haemorrhagic fevers (VHF) pose a significant threat to human health. In recent years, VHF outbreaks caused by Ebola, Marburg and Lassa viruses have caused substantial morbidity and mortality in West and Central Africa. In 2022, an Ebola disease outbreak in Uganda caused by Sudan virus resulted in 164 cases with 55 deaths. In 2023, a Marburg disease outbreak was confirmed in Equatorial Guinea and Tanzania resulting in over 49 confirmed or suspected cases; 41 of which were fatal. There are no clearly defined correlates of protection against these VHF, impeding targeted vaccine development. Any vaccine developed should therefore induce strong and preferably long-lasting humoral and cellular immunity against these viruses. Ideally this immunity should also cross-protect against viral variants, which are known to circulate in animal reservoirs and cause human disease. We have utilized two viral vectored vaccine platforms, an adenovirus (ChAdOx1) and Modified Vaccinia Ankara (MVA), to develop a multi-pathogen vaccine regime against three filoviruses (Ebola virus, Sudan virus, Marburg virus) and an arenavirus (Lassa virus). These platform technologies have consistently demonstrated the capability to induce robust cellular and humoral antigen-specific immunity in humans, most recently in the rollout of the licensed ChAdOx1-nCoV19/AZD1222. Here, we show that our multi-pathogen vaccines elicit strong cellular and humoral immunity, induce a diverse range of chemokines and cytokines, and most importantly, confers protection after lethal Ebola virus, Sudan virus and Marburg virus challenges in a small animal model.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Lassa Fever , Lassa virus , Marburg Virus Disease , Marburgvirus , Animals , Mice , Ebolavirus/immunology , Lassa virus/immunology , Marburgvirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/immunology , Lassa Fever/immunology , Lassa Fever/prevention & control , Marburg Virus Disease/immunology , Marburg Virus Disease/prevention & control , Viral Vaccines/immunology , Humans , Vaccination , Female , Antibodies, Viral/immunology , Immunogenicity, Vaccine , Ebola Vaccines/immunology
5.
Front Immunol ; 12: 774026, 2021.
Article in English | MEDLINE | ID: mdl-34777392

ABSTRACT

Marburg virus (MARV) is a member of the filovirus family that causes hemorrhagic disease with high case fatality rates. MARV is on the priority list of the World Health Organization for countermeasure development highlighting its potential impact on global public health. We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the MARV glycoprotein (VSV-MARV) and previously demonstrated uniform protection of nonhuman primates (NHPs) with a single dose. Here, we investigated the fast-acting potential of this vaccine by challenging NHPs with MARV 14, 7 or 3 days after a single dose vaccination with VSV-MARV. We found that 100% of the animals survived when vaccinated 7 or 14 days and 75% of the animal survived when vaccinated 3 days prior to lethal MARV challenge. Transcriptional analysis of whole blood samples indicated activation of B cells and antiviral defense after VSV-MARV vaccination. In the day -14 and -7 groups, limited transcriptional changes after challenge were observed with the exception of day 9 post-challenge in the day -7 group where we detected gene expression profiles indicative of a recall response. In the day -3 group, transcriptional analysis of samples from surviving NHPs revealed strong innate immune activation. In contrast, the animal that succumbed to disease in this group lacked signatures of antiviral immunity. In summary, our data demonstrate that the VSV-MARV is a fast-acting vaccine suitable for the use in emergency situations like disease outbreaks in Africa.


Subject(s)
Marburg Virus Disease/prevention & control , Marburgvirus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Chlorocebus aethiops , Cytokines/blood , Disease Models, Animal , Immunization , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Lymphocyte Activation , Marburg Virus Disease/blood , Marburg Virus Disease/immunology , Marburg Virus Disease/metabolism , Vaccination , Vero Cells , Vesiculovirus , Viral Load
6.
Front Immunol ; 12: 703986, 2021.
Article in English | MEDLINE | ID: mdl-34484200

ABSTRACT

Ebola (EBOV), Marburg (MARV) and Sudan (SUDV) viruses are the three filoviruses which have caused the most fatalities in humans. Transmission from animals into the human population typically causes outbreaks of limited scale in endemic regions. In contrast, the 2013-16 outbreak in several West African countries claimed more than 11,000 lives revealing the true epidemic potential of filoviruses. This is further emphasized by the difficulty seen with controlling the 2018-2020 outbreak of EBOV in the Democratic Republic of Congo (DRC), despite the availability of two emergency use-approved vaccines and several experimental therapeutics targeting EBOV. Moreover, there are currently no vaccine options to protect against the other epidemic filoviruses. Protection of a monovalent EBOV vaccine against other filoviruses has never been demonstrated in primate challenge studies substantiating a significant void in capability should a MARV or SUDV outbreak of similar magnitude occur. Herein we show progress on developing vaccines based on recombinant filovirus glycoproteins (GP) from EBOV, MARV and SUDV produced using the Drosophila S2 platform. The highly purified recombinant subunit vaccines formulated with CoVaccine HT™ adjuvant have not caused any safety concerns (no adverse reactions or clinical chemistry abnormalities) in preclinical testing. Candidate formulations elicit potent immune responses in mice, guinea pigs and non-human primates (NHPs) and consistently produce high antigen-specific IgG titers. Three doses of an EBOV candidate vaccine elicit full protection against lethal EBOV infection in the cynomolgus challenge model while one of four animals infected after only two doses showed delayed onset of Ebola Virus Disease (EVD) and eventually succumbed to infection while the other three animals survived challenge. The monovalent MARV or SUDV vaccine candidates completely protected cynomolgus macaques from infection with lethal doses of MARV or SUDV. It was further demonstrated that combinations of MARV or SUDV with the EBOV vaccine can be formulated yielding bivalent vaccines retaining full efficacy. The recombinant subunit vaccine platform should therefore allow the development of a safe and efficacious multivalent vaccine candidate for protection against Ebola, Marburg and Sudan Virus Disease.


Subject(s)
Ebola Vaccines/pharmacology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Marburg Virus Disease/prevention & control , Marburgvirus/immunology , Animals , Ebola Vaccines/genetics , Ebola Vaccines/immunology , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/immunology , Humans , Macaca fascicularis , Marburg Virus Disease/epidemiology , Marburg Virus Disease/genetics , Marburg Virus Disease/immunology , Marburgvirus/genetics , Vaccines, Synthetic
7.
Curr Biol ; 31(2): 257-270.e5, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33157026

ABSTRACT

Marburg virus (MARV) is among the most virulent pathogens of primates, including humans. Contributors to severe MARV disease include immune response suppression and inflammatory gene dysregulation ("cytokine storm"), leading to systemic damage and often death. Conversely, MARV causes little to no clinical disease in its reservoir host, the Egyptian rousette bat (ERB). Previous genomic and in vitro data suggest that a tolerant ERB immune response may underlie MARV avirulence, but no significant examination of this response in vivo yet exists. Here, using colony-bred ERBs inoculated with a bat isolate of MARV, we use species-specific antibodies and an immune gene probe array (NanoString) to temporally characterize the transcriptional host response at sites of MARV replication relevant to primate pathogenesis and immunity, including CD14+ monocytes/macrophages, critical immune response mediators, primary MARV targets, and skin at the inoculation site, where highest viral loads and initial engagement of antiviral defenses are expected. Our analysis shows that ERBs upregulate canonical antiviral genes typical of mammalian systems, such as ISG15, IFIT1, and OAS3, yet demonstrate a remarkable lack of significant induction of proinflammatory genes classically implicated in primate filoviral pathogenesis, including CCL8, FAS, and IL6. Together, these findings offer the first in vivo functional evidence for disease tolerance as an immunological mechanism by which the bat reservoir asymptomatically hosts MARV. More broadly, these data highlight factors determining disparate outcomes between reservoir and spillover hosts and defensive strategies likely utilized by bat hosts of other emerging pathogens, knowledge that may guide development of effective antiviral therapies.


Subject(s)
Chiroptera/immunology , Disease Reservoirs/virology , Immune Tolerance/immunology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Animals , Asymptomatic Infections , Chiroptera/blood , Chiroptera/genetics , Chiroptera/virology , Female , Gene Expression Regulation/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune Tolerance/genetics , Male , Marburg Virus Disease/virology , Monocytes/immunology
8.
Proc Natl Acad Sci U S A ; 117(49): 31142-31148, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229516

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.


Subject(s)
Antibodies, Viral/genetics , Complementarity Determining Regions/genetics , Marburg Virus Disease/immunology , Marburgvirus/immunology , 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
9.
Int J Infect Dis ; 99: 233-242, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32758690

ABSTRACT

OBJECTIVES: This article summarizes the countermeasures for Marburg virus disease, focusing on pathogenesis, clinical features and diagnostics. There is an emphasis on therapies and vaccines that have demonstrated, through their evaluation in nonhuman primates (NHPs) and/or in humans, potential for use in an emergency situation. METHODS: A standardized literature review was conducted on vaccines and treatments for Marburg virus disease, with a focus on human and nonhuman primate data published in the last five years. More detail on the methods that were used is summarized in a companion methods paper. RESULTS: The study identified six treatments and four vaccine platforms that have demonstrated, through their efficacy in NHPs, potential benefit for treating or preventing infection in humans. CONCLUSION: Succinct summaries of Marburg countermeasures are provided to give the busy clinician a head start in reviewing the literature if faced with a patient with Marburg virus disease. Links to other authoritative sources of information are also provided.


Subject(s)
Marburg Virus Disease/therapy , Animals , Humans , Marburg Virus Disease/immunology , Marburg Virus Disease/prevention & control , Marburg Virus Disease/virology , Marburgvirus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
10.
Cell Host Microbe ; 27(6): 976-991.e11, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32320678

ABSTRACT

Marburg virus (MARV) and Ebola virus (EBOV) belong to the family Filoviridae. MARV causes severe disease in humans with high fatality. We previously isolated a large panel of monoclonal antibodies (mAbs) from B cells of a human survivor with previous naturally acquired MARV infection. Here, we characterized functional properties of these mAbs and identified non-neutralizing mAbs targeting the glycoprotein (GP) 2 portion of the mucin-like domain (MLD) of MARV GP, termed the wing region. One mAb targeting the GP2 wing, MR228, showed therapeutic protection in mice and guinea pigs infected with MARV. The protection was mediated by the Fc fragment functions of MR228. Binding of another GP2 wing-specific non-neutralizing mAb, MR235, to MARV GP increased accessibility of epitopes in the receptor-binding site (RBS) for neutralizing mAbs, resulting in enhanced virus neutralization by these mAbs. These findings highlight an important role for non-neutralizing mAbs during natural human MARV infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Animals , Antibodies, Monoclonal/immunology , B-Lymphocytes , Chlorocebus aethiops , Disease Models, Animal , Ebolavirus/immunology , Epitopes/immunology , Female , Glycoproteins/immunology , Guinea Pigs , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Survivors , THP-1 Cells , Vero Cells , Viral Envelope Proteins/immunology
11.
Front Immunol ; 11: 435, 2020.
Article in English | MEDLINE | ID: mdl-32231668

ABSTRACT

Bats host a number of viruses that cause severe disease in humans without experiencing overt symptoms of disease themselves. While the mechanisms underlying this ability to avoid sickness are not known, deep sequencing studies of bat genomes have uncovered genetic adaptations that may have functional importance in the antiviral response of these animals. Egyptian rousette bats (Rousettus aegyptiacus) are the natural reservoir hosts of Marburg virus (MARV). In contrast to humans, these bats do not become sick when infected with MARV. A striking difference to the human genome is that Egyptian rousettes have an expanded repertoire of IFNW genes. To probe the biological implications of this expansion, we synthesized IFN-ω4 and IFN-ω9 proteins and tested their antiviral activity in Egyptian rousette cells. Both IFN-ω4 and IFN-ω9 showed antiviral activity against RNA viruses, including MARV, with IFN-ω9 being more efficient than IFN-ω4. Using RNA-Seq, we examined the transcriptional response induced by each protein. Although the sets of genes induced by the two IFNs were largely overlapping, IFN-ω9 induced a more rapid and intense response than did IFN-ω4. About 13% of genes induced by IFN-ω treatment are not found in the Interferome or other ISG databases, indicating that they may be uniquely IFN-responsive in this bat.


Subject(s)
Antiviral Agents/metabolism , Chiroptera/immunology , Disease Reservoirs/virology , Interferon Type I/metabolism , Marburg Virus Disease/immunology , Marburgvirus/immunology , Viral Zoonoses/immunology , Animals , Antibodies, Viral/blood , Chiroptera/virology , Host-Pathogen Interactions , Humans , Interferon Type I/genetics , Transcription, Genetic
12.
Sci Rep ; 10(1): 3071, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32080323

ABSTRACT

Postexposure immunization can prevent disease and reduce transmission following pathogen exposure. The rapid immunostimulatory properties of recombinant vesicular stomatitis virus (rVSV)-based vaccines make them suitable postexposure treatments against the filoviruses Ebola virus and Marburg virus (MARV); however, the mechanisms that drive this protection are undefined. Previously, we reported 60-75% survival of rhesus macaques treated with rVSV vectors expressing MARV glycoprotein (GP) 20-30 minutes after a low dose exposure to the most pathogenic variant of MARV, Angola. Survival in this model was linked to production of GP-specific antibodies and lower viral load. To confirm these results and potentially identify novel correlates of postexposure protection, we performed a similar experiment, but analyzed plasma cytokine levels, frequencies of immune cell subsets, and the transcriptional response to infection in peripheral blood. In surviving macaques (80-89%), we observed induction of genes mapping to antiviral and interferon-related pathways early after treatment and a higher percentage of T helper 1 (Th1) and NK cells. In contrast, the response of non-surviving macaques was characterized by hypercytokinemia; a T helper 2 signature; recruitment of low HLA-DR expressing monocytes and regulatory T-cells; and transcription of immune checkpoint (e.g., PD-1, LAG3) genes. These results suggest dysregulated immunoregulation is associated with poor prognosis, whereas early innate signaling and Th1-skewed immunity are important for survival.


Subject(s)
Marburg Virus Disease/immunology , Marburg Virus Disease/virology , Marburgvirus/immunology , Post-Exposure Prophylaxis , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Cytokines/blood , Cytotoxicity, Immunologic , Dose-Response Relationship, Immunologic , Down-Regulation/genetics , Female , Inflammation/blood , Inflammation/immunology , Interferons/genetics , Interferons/metabolism , Killer Cells, Natural/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , Male , Marburg Virus Disease/blood , Marburg Virus Disease/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombination, Genetic/genetics , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Transcriptome/genetics , Up-Regulation/genetics , Vesiculovirus/genetics , Viral Load/immunology
13.
Viruses ; 11(9)2019 08 24.
Article in English | MEDLINE | ID: mdl-31450611

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.


Subject(s)
Diagnostic Techniques and Procedures , Hemorrhagic Fever, Ebola , Information Services , RNA Virus Infections , Vaccines/standards , 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 , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/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 Virus Infections/diagnosis , RNA Virus Infections/immunology , RNA Virus Infections/prevention & control , 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
14.
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
15.
J Virol ; 93(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30567978

ABSTRACT

Marburg virus (MARV) is a filovirus related to Ebola virus (EBOV) associated with human hemorrhagic disease. Outbreaks are sporadic and severe, with a reported case mortality rate of upward of 88%. There is currently no antiviral or vaccine available. Given the sporadic nature of outbreaks, vaccines provide the best approach for long-term control of MARV in regions of endemicity. We have developed an inactivated rabies virus-vectored MARV vaccine (FILORAB3) to protect against Marburg virus disease. Immunogenicity studies in our labs have shown that a Th1-biased seroconversion to both rabies virus and MARV glycoproteins (GPs) is beneficial for protection in a preclinical murine model. As such, we adjuvanted FILORAB3 with glucopyranosyl lipid adjuvant (GLA), a Toll-like receptor 4 agonist, in a squalene-in-water emulsion. Across two different BALB/c mouse challenge models, we achieved 92% protection against murine-adapted Marburg virus (ma-MARV). Although our vaccine elicited strong MARV GP antibodies, it did not strongly induce neutralizing antibodies. Through both in vitro and in vivo approaches, we elucidated a critical role for NK cell-dependent antibody-mediated cellular cytotoxicity (ADCC) in vaccine-induced protection. Overall, these findings demonstrate that FILORAB3 is a promising vaccine candidate for Marburg virus disease.IMPORTANCE Marburg virus (MARV) is a virus similar to Ebola virus and also causes a hemorrhagic disease which is highly lethal. In contrast to EBOV, only a few vaccines have been developed against MARV, and researchers do not understand what kind of immune responses are required to protect from MARV. Here we show that antibodies directed against MARV after application of our vaccine protect in an animal system but fail to neutralize the virus in a widely used virus neutralization assay against MARV. This newly discovered activity needs to be considered more when analyzing MARV vaccines or infections.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Glycoproteins/immunology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Rabies virus/immunology , Rabies/immunology , Animals , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rabies Vaccines/immunology , Vaccination/methods , Vero Cells , Viral Vaccines/immunology
16.
J Infect Dis ; 219(11): 1716-1721, 2019 05 05.
Article in English | MEDLINE | ID: mdl-30590775

ABSTRACT

Although bats are increasingly being recognized as natural reservoir hosts of emerging zoonotic viruses, little is known about how they control and clear virus infection in the absence of clinical disease. Here, we test >50 convalescent sera from Egyptian rousette bats (ERBs) experimentally primed or prime-boosted with Marburg virus, Ebola virus, or Sosuga virus for the presence of virus-specific neutralizing antibodies, using infectious reporter viruses. After serum neutralization testing, we conclude that antibody-mediated virus neutralization does not contribute significantly to the control and clearance of Marburg virus, Ebola virus, or Sosuga virus infection in ERBs.


Subject(s)
Chiroptera/virology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Paramyxoviridae/immunology , Animals , Antibodies, Viral/immunology , Convalescence , Disease Reservoirs/virology , Egypt/epidemiology , Hemorrhagic Fever, Ebola/virology , Humans , Immunity, Humoral , Marburg Virus Disease/virology , Neutralization Tests
17.
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
18.
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
19.
Cell Host Microbe ; 24(3): 405-416.e3, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30173956

ABSTRACT

Sexual transmission of filoviruses was first reported in 1968 after an outbreak of Marburg virus (MARV) disease and recently caused flare-ups of Ebola virus disease in the 2013-2016 outbreak. How filoviruses establish testicular persistence and are shed in semen remain unknown. We discovered that persistent MARV infection of seminiferous tubules, an immune-privileged site that harbors sperm production, is a relatively common event in crab-eating macaques that survived infection after antiviral treatment. Persistence triggers severe testicular damage, including spermatogenic cell depletion and inflammatory cell invasion. MARV mainly persists in Sertoli cells, leading to breakdown of the blood-testis barrier formed by inter-Sertoli cell tight junctions. This disruption is accompanied by local infiltration of immunosuppressive CD4+Foxp3+ regulatory T cells. Our study elucidates cellular events associated with testicular persistence that may promote sexual transmission of filoviruses and suggests that targeting immunosuppression may be warranted to clear filovirus persistence in damaged immune-privileged sites.


Subject(s)
Marburg Virus Disease/virology , Marburgvirus/physiology , Primate Diseases/virology , Testis/virology , Animals , Macaca , Male , Marburg Virus Disease/immunology , Marburg Virus Disease/metabolism , Primate Diseases/immunology , Primate Diseases/metabolism , Sertoli Cells/metabolism , Sertoli Cells/virology , Survivors , T-Lymphocytes, Regulatory/immunology , Tight Junctions/metabolism , Tight Junctions/virology
20.
Cell Rep ; 24(7): 1802-1815.e5, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30110637

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , Antibody-Dependent Enhancement , Hemorrhagic Fever, Ebola/virology , Marburg Virus Disease/virology , 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
SELECTION OF CITATIONS
SEARCH DETAIL