Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Nat Commun ; 14(1): 6785, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37880247

ABSTRACT

Marburg and Ebola filoviruses are two of the deadliest infectious agents and several outbreaks have occurred in the last decades. Although several receptors and co-receptors have been reported for Ebola virus, key host factors remain to be elucidated. In this study, using a haploid cell screening platform, we identify the guanine nucleotide exchange factor CCZ1 as a key host factor in the early stage of filovirus replication. The critical role of CCZ1 for filovirus infections is validated in 3D primary human hepatocyte cultures and human blood-vessel organoids, both critical target sites for Ebola and Marburg virus tropism. Mechanistically, CCZ1 controls early to late endosomal trafficking of these viruses. In addition, we report that CCZ1 has a role in the endosomal trafficking of endocytosis-dependent SARS-CoV-2 infections, but not in infections by Lassa virus, which enters endo-lysosomal trafficking at the late endosome stage. Thus, we have identified an essential host pathway for filovirus infections in cell lines and engineered human target tissues. Inhibition of CCZ1 nearly completely abolishes Marburg and Ebola infections. Thus, targeting CCZ1 could potentially serve as a promising drug target for controlling infections caused by various viruses, such as SARS-CoV-2, Marburg, and Ebola.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Marburg Virus Disease , Marburgvirus , Vesicular Transport Proteins , Animals , Humans , Ebolavirus/metabolism , Lysosomes , Marburg Virus Disease/genetics , Marburg Virus Disease/metabolism , Marburgvirus/metabolism , Vesicular Transport Proteins/metabolism
2.
J Infect Dis ; 228(Suppl 7): S479-S487, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37119290

ABSTRACT

BACKGROUND: Our previous study demonstrated that the fruit bat (Yaeyama flying fox)-derived cell line FBKT1 showed preferential susceptibility to Ebola virus (EBOV), whereas the human cell line HEK293T was similarly susceptible to EBOV and Marburg virus (MARV). This was due to 3 amino acid differences of the endosomal receptor Niemann-Pick C1 (NPC1) between FBKT1 and HEK293T (ie, TET and SGA, respectively, at positions 425-427), as well as 2 amino acid differences at positions 87 and 142 of the viral glycoprotein (GP) between EBOV and MARV. METHODS/RESULTS: To understand the contribution of these amino acid differences to interactions between NPC1 and GP, we performed molecular dynamics simulations and binding free energy calculations. The average binding free energies of human NPC1 (hNPC1) and its mutant having TET at positions 425-427 (hNPC1/TET) were similar for the interaction with EBOV GP. In contrast, hNPC1/TET had a weaker interaction with MARV GP than wild-type hNPC1. As expected, substitutions of amino acid residues at 87 or 142 in EBOV and MARV GPs converted the binding affinity to hNPC1/TET. CONCLUSIONS: Our data provide structural and energetic insights for understanding potential differences in the GP-NPC1 interaction, which could influence the host tropism of EBOV and MARV.


Subject(s)
Chiroptera , Ebolavirus , Hemorrhagic Fever, Ebola , Marburgvirus , Animals , Humans , Niemann-Pick C1 Protein , Marburgvirus/metabolism , HEK293 Cells , Virus Internalization , Glycoproteins/metabolism , Ebolavirus/metabolism , Amino Acids
3.
Autophagy ; 19(10): 2809-2810, 2023 10.
Article in English | MEDLINE | ID: mdl-36763514

ABSTRACT

Ebola virus (EBOV) and Marburg virus (MARV) are zoonotic, virulent pathogens that cause sporadic and global outbreaks of severe hemorrhagic fever. Reemergence of these filoviruses remains a global public health threat, highlighting the need for novel countermeasures to control and treat future disease outbreaks. The EBOV VP40 matrix protein drives virion assembly and egress. We recently reported that BAG3 and HSPA/HSP70, two central components of chaperone-assisted selective autophagy (CASA), target VP40 for autophagic sequestration and degradation, thereby inhibiting virus egress and spread. In addition, we found that expression of the EBOV glycoprotein (GP) activates MTORC1, the gateway regulator of autophagy. Notably, pharmacological suppression of MTORC1 signaling by rapamycin activates autophagy and blocks filovirus egress. These findings highlight the MTORC1-CASA axis as a regulator of filovirus egress and suggest new opportunities for antiviral development and intervention.


Subject(s)
Ebolavirus , Marburgvirus , Autophagy , Marburgvirus/metabolism
4.
Microscopy (Oxf) ; 72(3): 178-190, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-36242583

ABSTRACT

Filoviruses are filamentous enveloped viruses belonging to the family Filoviridae, in the order Mononegavirales. Some filovirus members, such as Ebola virus and Marburg virus, cause severe hemorrhagic fever in humans and non-human primates. The filovirus ribonucleoprotein complex, called the nucleocapsid, forms a double-layered helical structure in which a non-segmented, single-stranded, negative-sense RNA genome is encapsidated by the nucleoprotein (NP), viral protein 35 (VP35), VP24, VP30 and RNA-dependent RNA polymerase (L). The inner layer consists of the helical NP-RNA complex, acting as a scaffold for the binding of VP35 and VP24 that constitute the outer layer. Recent structural studies using cryo-electron microscopy have advanced our understanding of the molecular mechanism of filovirus nucleocapsid formation. Here, we review the key characteristics of the Ebola virus and Marburg virus nucleocapsid structures, highlighting the similarities and differences between the two viruses. In particular, we focus on the structure of the helical NP-RNA complex, the RNA binding mechanism and the NP-NP interactions in the helix. The structural analyses reveal a possible mechanism of nucleocapsid assembly and provide potential targets for the anti-filovirus drug design.


Subject(s)
Ebolavirus , Marburgvirus , Animals , Cryoelectron Microscopy , Nucleocapsid/chemistry , Nucleocapsid/metabolism , Ebolavirus/chemistry , Ebolavirus/metabolism , Marburgvirus/chemistry , Marburgvirus/metabolism , Viral Proteins/analysis , Viral Proteins/chemistry , Viral Proteins/metabolism , RNA/analysis , RNA/metabolism
5.
J Virol ; 96(6): e0202621, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35107375

ABSTRACT

Ebola virus (EBOV) and Marburg virus (MARV) continue to emerge and cause severe hemorrhagic disease in humans. A comprehensive understanding of the filovirus-host interplay will be crucial for identifying and developing antiviral strategies. The filoviral VP40 matrix protein drives virion assembly and egress, in part by recruiting specific WW domain-containing host interactors via its conserved PPxY late (L) domain motif to positively regulate virus egress and spread. In contrast to these positive regulators of virus budding, a growing list of WW domain-containing interactors that negatively regulate virus egress and spread have been identified, including BAG3, YAP/TAZ, and WWOX. In addition to host WW domain regulators of virus budding, host PPxY-containing proteins also contribute to regulating this late stage of filovirus replication. For example, angiomotin (AMOT) is a multi-PPxY-containing host protein that functionally interacts with many of the same WW domain-containing proteins that regulate virus egress and spread. In this report, we demonstrate that host WWOX, which negatively regulates egress of VP40 virus-like particles (VLPs) and recombinant vesicular stomatitis virus (VSV) M40 virus, interacts with and suppresses the expression of AMOT. We found that WWOX disrupts AMOT's scaffold-like tubular distribution and reduces AMOT localization at the plasma membrane via lysosomal degradation. In sum, our findings reveal an indirect and novel mechanism by which modular PPxY-WW domain interactions between AMOT and WWOX regulate PPxY-mediated egress of filovirus VP40 VLPs. A better understanding of this modular network and competitive nature of protein-protein interactions will help to identify new antiviral targets and therapeutic strategies. IMPORTANCE Filoviruses (Ebola virus [EBOV] and Marburg virus [MARV]) are zoonotic, emerging pathogens that cause outbreaks of severe hemorrhagic fever in humans. A fundamental understanding of the virus-host interface is critical for understanding the biology of these viruses and for developing future strategies for therapeutic intervention. Here, we reveal a novel mechanism by which host proteins WWOX and AMOTp130 interact with each other and with the filovirus matrix protein VP40 to regulate VP40-mediated egress of virus-like particles (VLPs). Our results highlight the biological impact of competitive interplay of modular virus-host interactions on both the virus life cycle and the host cell.


Subject(s)
Ebolavirus , Marburgvirus , WW Domain-Containing Oxidoreductase , Angiomotins/metabolism , Ebolavirus/physiology , Humans , Marburgvirus/metabolism , Viral Matrix Proteins/metabolism , Virus Release/physiology , WW Domain-Containing Oxidoreductase/metabolism
6.
J Virol ; 95(19): e0065221, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34346762

ABSTRACT

The filovirus family includes deadly pathogens such as Ebola virus (EBOV) and Marburg virus (MARV). A substantial portion of filovirus genomes encode 5' and 3' untranslated regions (UTRs) of viral mRNAs. Select viral genomic RNA sequences corresponding to 3' UTRs are prone to editing by adenosine deaminase acting on RNA 1 (ADAR1). A reporter mRNA approach, in which different 5' or 3' UTRs were inserted into luciferase-encoding mRNAs, demonstrates that MARV 3' UTRs yield different levels of reporter gene expression, suggesting modulation of translation. The modulation occurs in cells unable to produce microRNAs (miRNAs) and can be recapitulated in a MARV minigenome assay. Deletion mutants identified negative regulatory regions at the ends of the MARV nucleoprotein (NP) and large protein (L) 3' UTRs. Apparent ADAR1 editing mutants were previously identified within the MARV NP 3' UTR. Introduction of these changes into the MARV nucleoprotein (NP) 3' UTR or deletion of the region targeted for editing enhances translation, as indicated by reporter assays and polysome analysis. In addition, the parental NP 3' UTR, but not the edited or deletion mutant NP 3' UTRs, induces a type I interferon (IFN) response upon transfection into cells. Because some EBOV isolates from the West Africa outbreak exhibited ADAR1 editing of the viral protein of 40 kDa (VP40) 3' UTR, VP40 3' UTRs with parental and edited sequences were similarly assayed. The EBOV VP40 3' UTR edits also enhanced translation, but neither the wild-type nor the edited 3' UTRs induced IFN. These findings implicate filoviral mRNA 3' UTRs as negative regulators of translation that can be inactivated by innate immune responses that induce ADAR1. IMPORTANCE UTRs comprise a large percentage of filovirus genomes and are apparent targets of editing by ADAR1, an enzyme with pro- and antiviral activities. However, the functional significance of the UTRs and ADAR1 editing has been uncertain. This study demonstrates that MARV and EBOV 3' UTRs can modulate translation, in some cases negatively. ADAR1 editing or deletion of select regions within the translation suppressing 3' UTRs relieves the negative effects of the UTRs. These data indicate that filovirus 3' UTRs contain translation regulatory elements that are modulated by activation of ADAR1, suggesting a complex interplay between filovirus gene expression and innate immunity.


Subject(s)
3' Untranslated Regions , Adenosine Deaminase/metabolism , Ebolavirus/genetics , Marburgvirus/genetics , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Animals , Cell Line , Ebolavirus/metabolism , Genes, Reporter , Humans , Interferon Type I/biosynthesis , Marburgvirus/metabolism , MicroRNAs/genetics , Mutation , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Polyribosomes/metabolism , RNA Editing , RNA, Messenger/genetics , RNA, Messenger/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
7.
Cell Rep ; 30(13): 4540-4550.e3, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32234486

ABSTRACT

Ebola virus causes severe hemorrhagic fever, often leading to death in humans. The trimeric fusion glycoprotein (GP) is the sole target for neutralizing antibodies and is the major focus of vaccine development. Soluble GP ectodomains are unstable and mostly monomeric when not fused to a heterologous trimerization domain. Here, we report structure-based designs of Ebola and Marburg GP trimers based on a stabilizing mutation in the hinge loop in refolding region 1 and substitution of a partially buried charge at the interface of the GP1 and GP2 subunits. The combined substitutions (T577P and K588F) substantially increased trimer expression for Ebola GP proteins. We determined the crystal structure of stabilized GP from the Makona Zaire ebolavirus strain without a trimerization domain or complexed ligand. The structure reveals that the stabilized GP adopts the same trimeric prefusion conformation, provides insight into triggering of GP conformational changes, and should inform future filovirus vaccine development.


Subject(s)
Filoviridae/metabolism , Glycoproteins/chemistry , Protein Multimerization , Amino Acid Substitution , Cell Line , Crystallography, X-Ray , Ebolavirus/metabolism , Glycoproteins/genetics , Humans , Marburgvirus/metabolism , Models, Molecular , Mutation/genetics , Perfusion , Protein Domains , Protein Stability , Structure-Activity Relationship
8.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31852785

ABSTRACT

The question as to whether RNA viruses produce bona fide microRNAs (miRNAs) during infection has been the focus of intense research and debate. Recently, several groups using computational prediction methods have independently reported possible miRNA candidates produced by Ebola virus (EBOV). Additionally, efforts to detect these predicted RNA products in samples from infected animals and humans have produced positive results. However, these studies and their conclusions are predicated on the assumption that these RNA products are actually processed through, and function within, the miRNA pathway. In the present study, we performed the first rigorous assessment of the ability of filoviruses to produce miRNA products during infection of both human and bat cells. Using next-generation sequencing, we detected several candidate miRNAs from both EBOV and the closely related Marburg virus (MARV). Focusing our validation efforts on EBOV, we found evidence contrary to the idea that these small RNA products function as miRNAs. The results of our study are important because they highlight the potential pitfalls of relying on computational methods alone for virus miRNA discovery.IMPORTANCE Here, we report the discovery, via deep sequencing, of numerous noncoding RNAs (ncRNAs) derived from both EBOV and MARV during infection of both bat and human cell lines. In addition to identifying several novel ncRNAs from both viruses, we identified two EBOV ncRNAs in our sequencing data that were near-matches to computationally predicted viral miRNAs reported in the literature. Using molecular and immunological techniques, we assessed the potential of EBOV ncRNAs to function as viral miRNAs. Importantly, we found little evidence supporting this hypothesis. Our work is significant because it represents the first rigorous assessment of the potential for EBOV to encode viral miRNAs and provides evidence contrary to the existing paradigm regarding the biological role of computationally predicted EBOV ncRNAs. Moreover, our work highlights further avenues of research regarding the nature and function of EBOV ncRNAs.


Subject(s)
Ebolavirus/metabolism , MicroRNAs/metabolism , RNA Interference , RNA, Viral/metabolism , Animals , Cell Line , Chiroptera , Ebolavirus/genetics , Humans , Marburgvirus/genetics , Marburgvirus/metabolism , MicroRNAs/genetics , RNA, Viral/genetics
9.
Virology ; 532: 22-29, 2019 06.
Article in English | MEDLINE | ID: mdl-30999160

ABSTRACT

Priming of the viral glycoprotein (GP) by the cellular proteases cathepsin B and L (CatB, CatL) is believed to be essential for cell entry of filoviruses. However, pseudotyping systems that predominantly produce non-filamentous particles have frequently been used to prove this concept. Here, we report that GP-mediated entry of retroviral-, rhabdoviral and filoviral particles depends on CatB/CatL activity and that this effect is cell line-independent. Moreover, we show that the human cell line Calu-3, which expresses low amounts of CatL, is largely resistant to entry driven by diverse filovirus GPs. Finally, we demonstrate that Calu-3 cell entry mediated by certain filovirus GPs can be rescued upon directed expression of CatL or DC-SIGN. Our results identify Calu-3 cells as largely resistant to filovirus GP-driven entry and demonstrate that entry is limited at the stage of virion attachment and GP priming.


Subject(s)
Cathepsin L/genetics , Cell Adhesion Molecules/genetics , Ebolavirus/genetics , Epithelial Cells/immunology , Lectins, C-Type/genetics , Receptors, Cell Surface/genetics , Viral Proteins/genetics , A549 Cells , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cathepsin B/immunology , Cathepsin B/metabolism , Cathepsin L/antagonists & inhibitors , Cathepsin L/immunology , Cathepsin L/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Ebolavirus/growth & development , Ebolavirus/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression Regulation , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Leucine/analogs & derivatives , Leucine/pharmacology , Marburgvirus/genetics , Marburgvirus/growth & development , Marburgvirus/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signal Transduction , Vero Cells , Vesiculovirus/genetics , Vesiculovirus/growth & development , Vesiculovirus/metabolism , Viral Proteins/metabolism , Virion/genetics , Virion/growth & development , Virion/metabolism , Virus Internalization/drug effects
10.
J Infect Dis ; 218(suppl_5): S403-S408, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30165526

ABSTRACT

Protein kinase R (PKR) is a key antiviral protein involved in sensing and restricting viral infections. Here we analyzed the ability of Marburg virus (MARV) viral protein 35 (VP35) to inhibit PKR activation in human and bat cells. Similar to the related Ebola and Lloviu viruses, MARV VP35 was able to inhibit PKR activation in 293T cells. In contrast, we found that MARV VP35 did not inhibit human or bat PKR activation in human glioblastoma U-251-MG cells or a Rousettus aegyptiacus cell line. Additional experiments revealed that PACT, a known PKR regulator, was insufficient to rescue the ability of VP35 to inhibit PKR activation in these cells. Taken together, this study indicates that the ability of VP35 to inhibit PKR is cell type specific, potentially explaining discrepancies between the ability of filoviruses to potently block innate immune responses, and the high levels of interferon and interferon-stimulated genes observed in filovirus patients.


Subject(s)
Marburgvirus/metabolism , Protein Kinases/metabolism , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Animals , Cell Line , Chiroptera , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/immunology , Marburg Virus Disease/immunology , Marburg Virus Disease/metabolism , Marburgvirus/immunology , Protein Kinases/immunology , Viral Proteins/immunology , Viral Regulatory and Accessory Proteins/immunology
11.
J Infect Dis ; 218(suppl_5): S318-S326, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30165666

ABSTRACT

The single surface glycoprotein (GP) of filoviruses is indispensable for recognition of its cellular receptor and infection of target cells. To study the intracellular trafficking of GP by using live-cell imaging, the mucin-like domain of Marburg virus (MARV) GP was replaced by the fluorophore mCherry (GP∆MLD_mCherry). Intracellular distribution, surface transport, and recruitment of GP∆MLD_mCherry into virus-like particles were similar to observations for wild-type GP. Using reverse genetics, we generated a recombinant MARV expressing GP∆MLD_mCherry (recMARV MARVGP∆MLD_mCherry). Time-lapse microscopy of recMARV MARVGP∆MLD_mCherry-infected cells revealed that GP∆MLD_mCherry-positive vesicles were transported to the cell surface in a tubulin-dependent manner. Moreover, dual-color live-cell imaging revealed cotransport of GPΔMLD_mCherry and VP40 and their colocalization at the plasma membrane. In this proof-of-concept study we showed that the newly developed GP∆MLD_mCherry is a promising tool to elucidate intracellular trafficking and assembly pathways of MARV.


Subject(s)
Fluorescent Dyes/administration & dosage , Glycoproteins/metabolism , Marburgvirus/metabolism , Marburgvirus/physiology , Protein Transport/physiology , Virus Assembly/physiology , Virus Release/physiology , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/physiology , Cell Membrane/virology , HEK293 Cells , Humans
12.
Cell Host Microbe ; 23(1): 101-109.e4, 2018 Jan 10.
Article in English | MEDLINE | ID: mdl-29324225

ABSTRACT

Since their first identification 50 years ago, marburgviruses have emerged several times, with 83%-90% lethality in the largest outbreaks. Although no vaccines or therapeutics are available for human use, the human antibody MR191 provides complete protection in non-human primates when delivered several days after inoculation of a lethal marburgvirus dose. The detailed neutralization mechanism of MR191 remains outstanding. Here we present a 3.2 Å crystal structure of MR191 complexed with a trimeric marburgvirus surface glycoprotein (GP). MR191 neutralizes by occupying the conserved receptor-binding site and competing with the host receptor Niemann-Pick C1. The structure illuminates previously disordered regions of GP including the stalk, fusion loop, CX6CC switch, and an N-terminal region of GP2 that wraps about the outside of GP1 to anchor a marburgvirus-specific "wing" antibody epitope. Virus escape mutations mapped far outside the MR191 receptor-binding site footprint suggest a role for these other regions in the GP quaternary structure.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Marburgvirus/immunology , Receptors, Virus/immunology , Receptors, Virus/ultrastructure , Viral Fusion Proteins/immunology , Viral Fusion Proteins/ultrastructure , Agrobacterium tumefaciens , Animals , Antibodies, Monoclonal/ultrastructure , Binding Sites/immunology , Carrier Proteins/immunology , Cell Line , Chlorocebus aethiops , Crystallography, X-Ray , Drosophila melanogaster , Humans , Intracellular Signaling Peptides and Proteins , Marburgvirus/metabolism , Membrane Glycoproteins/immunology , Niemann-Pick C1 Protein , Nicotiana , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virus Attachment
13.
Methods Mol Biol ; 1604: 209-215, 2018.
Article in English | MEDLINE | ID: mdl-28986836

ABSTRACT

Independent expression of the VP40 or Z matrix proteins of filoviruses (marburgviruses and ebolaviruses) and arenaviruses (Lassa fever and Junín), respectively, gives rise to the production and release of virus-like particles (VLPs) that are morphologically identical to infectious virions. We can detect and quantify VLP production and egress in mammalian cells by transient transfection, SDS-PAGE, Western blotting, and live cell imaging techniques such as total internal reflection fluorescence (TIRF) microscopy. Since the VLP budding assay accurately mimics budding of infectious virus, this BSL-2 assay is safe and useful for the interrogation of both viral and host determinants required for budding and can be used as an initial screen to identify and validate small molecule inhibitors of virus release and spread.


Subject(s)
Hemorrhagic Fever, Ebola/metabolism , Virus Release/physiology , Animals , Arenavirus/genetics , Arenavirus/metabolism , Blotting, Western , Ebolavirus/genetics , Ebolavirus/metabolism , Electrophoresis, Polyacrylamide Gel , Filoviridae/genetics , Filoviridae/metabolism , Hemorrhagic Fever, Ebola/genetics , Humans , Junin virus/genetics , Junin virus/metabolism , Lassa Fever/virology , Marburgvirus/genetics , Marburgvirus/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virus Release/genetics
14.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142131

ABSTRACT

Previous studies demonstrated that a single intramuscular (i.m.) dose of an attenuated recombinant vesicular stomatitis virus (rVSV) vector (VesiculoVax vector platform; rVSV-N4CT1) expressing the glycoprotein (GP) from the Mayinga strain of Zaire ebolavirus (EBOV) protected nonhuman primates (NHPs) from lethal challenge with EBOV strains Kikwit and Makona. Here, we studied the immunogenicities of an expanded range of attenuated rVSV vectors expressing filovirus GP in mice. Based on data from those studies, an optimal attenuated trivalent rVSV vector formulation was identified that included rVSV vectors expressing EBOV, Sudan ebolavirus (SUDV), and the Angola strain of Marburg marburgvirus (MARV) GPs. NHPs were vaccinated with a single dose of the trivalent formulation, followed by lethal challenge 28 days later with each of the three corresponding filoviruses. At day 14 postvaccination, a serum IgG response specific for all three GPs was detected in all the vaccinated macaques. A modest and balanced cell-mediated immune response specific for each GP was also detected in a majority of the vaccinated macaques. No matter the level of total GP-specific immune response detected postvaccination, all the vaccinated macaques were protected from disease and death following lethal challenge with each of the three filoviruses. These findings indicate that vaccination with a single dose of attenuated rVSV-N4CT1 vectors each expressing a single filovirus GP may provide protection against the filoviruses most commonly responsible for outbreaks of hemorrhagic fever in sub-Saharan Africa.IMPORTANCE The West African Ebola virus Zaire outbreak in 2013 showed that the disease was not only a regional concern, but a worldwide problem, and highlighted the need for a safe and efficacious vaccine to be administered to the populace. However, other endemic pathogens, like Ebola virus Sudan and Marburg, also pose an important health risk to the public and therefore require development of a vaccine prior to the occurrence of an outbreak. The significance of our research was the development of a blended trivalent filovirus vaccine that elicited a balanced immune response when administered as a single dose and provided complete protection against a lethal challenge with all three filovirus pathogens.


Subject(s)
Ebolavirus/metabolism , Glycoproteins/metabolism , Hemorrhagic Fever, Ebola/prevention & control , Marburg Virus Disease/prevention & control , Marburgvirus/metabolism , Vesiculovirus/genetics , Viral Vaccines/administration & dosage , Animals , Antibodies, Viral/metabolism , Ebolavirus/immunology , Glycoproteins/genetics , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/immunology , Immunoglobulin G/metabolism , Injections, Intramuscular , Macaca fascicularis , Marburg Virus Disease/immunology , Marburgvirus/immunology , Mice , Vaccination , Vaccines, Attenuated , Vaccines, Synthetic , Vesiculovirus/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Viral Vaccines/immunology
15.
Antiviral Res ; 141: 48-61, 2017 05.
Article in English | MEDLINE | ID: mdl-28192094

ABSTRACT

Filoviruses are important pathogens that cause severe and often fatal hemorrhagic fever in humans, for which no approved vaccines and antiviral treatments are yet available. In an earlier article (Martin et al., Antiviral Research, 2016), we reviewed the role of the filovirus surface glycoprotein in replication and as a target for drugs and vaccines. In this review, we focus on recent findings on the filovirus replication machinery and how they could be used for the identification of new therapeutic targets and the development of new antiviral compounds. First, we summarize the recent structural and functional advances on the molecules involved in filovirus replication/transcription cycle, particularly the NP, VP30, VP35 proteins, and the "large" protein L, which harbors the RNA-dependent RNA polymerase (RdRp) and mRNA capping activities. These proteins are essential for viral mRNA synthesis and genome replication, and consequently they constitute attractive targets for drug design. We then describe how these insights into filovirus replication mechanisms and the structure/function characterization of the involved proteins have led to the development of new and innovative antiviral strategies that may help reduce the filovirus disease case fatality rate through post-exposure or prophylactic treatments.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Filoviridae/drug effects , Filoviridae/physiology , Viral Proteins/metabolism , Virus Replication , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Drug Design , Ebolavirus/chemistry , Ebolavirus/drug effects , Ebolavirus/metabolism , Filoviridae Infections/drug therapy , Marburgvirus/chemistry , Marburgvirus/drug effects , Marburgvirus/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/isolation & purification , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism
16.
J Biol Chem ; 292(15): 6108-6122, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28167534

ABSTRACT

Marburg virus (MARV) is a lipid-enveloped virus from the Filoviridae family containing a negative sense RNA genome. One of the seven MARV genes encodes the matrix protein VP40, which forms a matrix layer beneath the plasma membrane inner leaflet to facilitate budding from the host cell. MARV VP40 (mVP40) has been shown to be a dimeric peripheral protein with a broad and flat basic surface that can associate with anionic phospholipids such as phosphatidylserine. Although a number of mVP40 cationic residues have been shown to facilitate binding to membranes containing anionic lipids, much less is known on how mVP40 assembles to form the matrix layer following membrane binding. Here we have used hydrogen/deuterium exchange (HDX) mass spectrometry to determine the solvent accessibility of mVP40 residues in the absence and presence of phosphatidylserine and phosphatidylinositol 4,5-bisphosphate. HDX analysis demonstrates that two basic loops in the mVP40 C-terminal domain make important contributions to anionic membrane binding and also reveals a potential oligomerization interface in the C-terminal domain as well as a conserved oligomerization interface in the mVP40 N-terminal domain. Lipid binding assays confirm the role of the two basic patches elucidated with HD/X measurements, whereas molecular dynamics simulations and membrane insertion measurements complement these studies to demonstrate that mVP40 does not appreciably insert into the hydrocarbon region of anionic membranes in contrast to the matrix protein from Ebola virus. Taken together, we propose a model by which association of the mVP40 dimer with the anionic plasma membrane facilitates assembly of mVP40 oligomers.


Subject(s)
Marburgvirus/chemistry , Models, Chemical , Phosphatidylcholines/chemistry , Phosphatidylserines/chemistry , Protein Multimerization , Viral Matrix Proteins/chemistry , Deuterium Exchange Measurement , Marburgvirus/genetics , Marburgvirus/metabolism , Mass Spectrometry , Protein Structure, Quaternary , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
17.
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
18.
Virology ; 501: 147-165, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27930961

ABSTRACT

Filoviruses are highly lethal in humans and nonhuman primates, likely due to potent antagonism of host interferon (IFN) responses early in infection. Filoviral protein VP35 is implicated as the major IFN induction antagonist, while Ebola virus (EBOV) VP24 or Marburg virus (MARV) VP40 are known to block downstream IFN signaling. Despite progress elucidating EBOV and MARV antagonist function, those for most other filoviruses, including Reston (RESTV), Sudan (SUDV), Taï Forest (TAFV), Bundibugyo (BDBV) and Ravn (RAVV) viruses, remain largely neglected. Thus, using standardized vectors and reporter assays, we characterized activities by each IFN antagonist from all known ebolavirus and marburgvirus species side-by-side. We uncover noncanonical suppression of IFN induction by ebolavirus VP24, differing potencies by MARV and RAVV proteins, and intriguingly, weaker antagonism by VP24 of RESTV. These underlying molecular explanations for differential virulence in humans could guide future investigations of more-neglected filoviruses as well as treatment and vaccine studies.


Subject(s)
Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/virology , Interferons/antagonists & inhibitors , Marburg Virus Disease/virology , Marburgvirus/metabolism , Viral Proteins/metabolism , Animals , Ebolavirus/genetics , Genes, Reporter , Host-Pathogen Interactions , Humans , Interferons/metabolism , Marburgvirus/genetics , Viral Proteins/genetics
19.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27847355

ABSTRACT

Marburg virus (MARV) is a highly pathogenic filovirus that is classified in a genus distinct from that of Ebola virus (EBOV) (genera Marburgvirus and Ebolavirus, respectively). Both viruses produce a multifunctional protein termed VP35, which acts as a polymerase cofactor, a viral protein chaperone, and an antagonist of the innate immune response. VP35 contains a central oligomerization domain with a predicted coiled-coil motif. This domain has been shown to be essential for RNA polymerase function. Here we present crystal structures of the MARV VP35 oligomerization domain. These structures and accompanying biophysical characterization suggest that MARV VP35 is a trimer. In contrast, EBOV VP35 is likely a tetramer in solution. Differences in the oligomeric state of this protein may explain mechanistic differences in replication and immune evasion observed for MARV and EBOV. IMPORTANCE: Marburg virus can cause severe disease, with up to 90% human lethality. Its genome is concise, only producing seven proteins. One of the proteins, VP35, is essential for replication of the viral genome and for evasion of host immune responses. VP35 oligomerizes (self-assembles) in order to function, yet the structure by which it assembles has not been visualized. Here we present two crystal structures of this oligomerization domain. In both structures, three copies of VP35 twist about each other to form a coiled coil. This trimeric assembly is in contrast to tetrameric predictions for VP35 of Ebola virus and to known structures of homologous proteins in the measles, mumps, and Nipah viruses. Distinct oligomeric states of the Marburg and Ebola virus VP35 proteins may explain differences between them in polymerase function and immune evasion. These findings may provide a more accurate understanding of the mechanisms governing VP35's functions and inform the design of therapeutics.


Subject(s)
Marburgvirus/metabolism , Models, Molecular , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Viral Regulatory and Accessory Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Protein Binding , Protein Stability , Thermodynamics , Viral Regulatory and Accessory Proteins/metabolism
20.
Sci Rep ; 6: 34589, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713552

ABSTRACT

The unprecedented outbreak of Ebola in West Africa resulted in over 28,000 cases and 11,000 deaths, underlining the need for a better understanding of the biology of this highly pathogenic virus to develop specific counter strategies. Two filoviruses, the Ebola and Marburg viruses, result in a severe and often fatal infection in humans. However, bats are natural hosts and survive filovirus infections without obvious symptoms. The molecular basis of this striking difference in the response to filovirus infections is not well understood. We report a systematic overview of differentially expressed genes, activity motifs and pathways in human and bat cells infected with the Ebola and Marburg viruses, and we demonstrate that the replication of filoviruses is more rapid in human cells than in bat cells. We also found that the most strongly regulated genes upon filovirus infection are chemokine ligands and transcription factors. We observed a strong induction of the JAK/STAT pathway, of several genes encoding inhibitors of MAP kinases (DUSP genes) and of PPP1R15A, which is involved in ER stress-induced cell death. We used comparative transcriptomics to provide a data resource that can be used to identify cellular responses that might allow bats to survive filovirus infections.


Subject(s)
Ebolavirus/metabolism , Gene Expression Regulation , Hemorrhagic Fever, Ebola/metabolism , Marburg Virus Disease/metabolism , Marburgvirus/metabolism , Signal Transduction , Transcription, Genetic , Animals , Cell Line, Tumor , Chiroptera , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...