Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 11(6): e0237323, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37888996

ABSTRACT

IMPORTANCE: Filoviruses are the causative agents of severe and often fatal hemorrhagic disease in humans. Menglà virus (MLAV) is a recently reported filovirus, isolated from fruit bats that is capable to replicate in human cells, representing a potential risk for human health. An in-depth structural and functional knowledge of MLAV proteins is an essential step for antiviral research on this virus that can also be extended to other emerging filoviruses. In this study, we determined the first crystal structures of the C-terminal domain (CTD) of the MLAV nucleoprotein (NP), showing important similarities to the equivalent domain in MARV. The structural data also show that the NP CTD has the ability to form large helical oligomers that may participate in the control of cytoplasmic inclusion body formation during viral replication.


Subject(s)
Ebolavirus , Filoviridae , Humans , Nucleoproteins/chemistry , Filoviridae/chemistry , Filoviridae/metabolism , Viral Proteins/metabolism
2.
Viruses ; 13(5)2021 05 14.
Article in English | MEDLINE | ID: mdl-34069246

ABSTRACT

Filoviruses, including marburgviruses and ebolaviruses, have a single transmembrane glycoprotein (GP) that facilitates their entry into cells. During entry, GP needs to be cleaved by host proteases to expose the receptor-binding site that binds to the endosomal receptor Niemann-Pick C1 (NPC1) protein. The crystal structure analysis of the cleaved GP (GPcl) of Ebola virus (EBOV) in complex with human NPC1 has demonstrated that NPC1 has two protruding loops (loops 1 and 2), which engage a hydrophobic pocket on the head of EBOV GPcl. However, the molecular interactions between NPC1 and the GPcl of other filoviruses remain unexplored. In the present study, we performed molecular modeling and molecular dynamics simulations of NPC1 complexed with GPcls of two ebolaviruses, EBOV and Sudan virus (SUDV), and one marburgvirus, Ravn virus (RAVV). Similar binding structures were observed in the GPcl-NPC1 complexes of EBOV and SUDV, which differed from that of RAVV. Specifically, in the RAVV GPcl-NPC1 complex, the tip of loop 2 was closer to the pocket edge comprising residues at positions 79-88 of GPcl; the root of loop 1 was predicted to interact with P116 and Q144 of GPcl. Furthermore, in the SUDV GPcl-NPC1 complex, the tip of loop 2 was slightly closer to the residue at position 141 than those in the EBOV and RAVV GPcl-NPC1 complexes. These structural differences may affect the size and/or shape of the receptor-binding pocket of GPcl. Our structural models could provide useful information for improving our understanding the differences in host preference among filoviruses as well as contributing to structure-based drug design.


Subject(s)
Filoviridae , Models, Molecular , Niemann-Pick C1 Protein/chemistry , Niemann-Pick C1 Protein/metabolism , Protein Conformation , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Binding Sites , Filoviridae/metabolism , Protein Binding , Structure-Activity Relationship
3.
Arch Virol ; 165(10): 2165-2176, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32740830

ABSTRACT

The PI3K/Akt signalling pathway is a crucial signalling cascade that regulates transcription, protein translation, cell growth, proliferation, cell survival, and metabolism. During viral infection, viruses exploit a variety of cellular pathways, including the well-known PI3K/Akt signalling pathway. Conversely, cells rely on this pathway to stimulate an antiviral response. The PI3K/Akt pathway is manipulated by a number of viruses, including DNA and RNA viruses and retroviruses. The aim of this review is to provide up-to-date information about the role of the PI3K-Akt pathway in infection with members of five different families of negative-sense ssRNA viruses. This pathway is hijacked for viral entry, regulation of endocytosis, suppression of premature apoptosis, viral protein expression, and replication. Although less common, the PI3K/Akt pathway can be downregulated as an immunomodulatory strategy or as a mechanism for inducing autophagy. Moreover, the cell activates this pathway as an antiviral strategy for interferon and cytokine production, among other strategies. Here, we present new data concerning the role of this pathway in infection with the paramyxovirus Newcastle disease virus (NDV). Our data seem to indicate that NDV uses the PI3K/Akt pathway to delay cell death and increase cell survival as a means of improving its replication. The interference of negative-sense ssRNA viruses with this essential pathway might have implications for the development of antiviral therapies.


Subject(s)
Gene Expression Regulation , Host-Pathogen Interactions/genetics , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA Virus Infections/genetics , Apoptosis/genetics , Autophagy/genetics , Autophagy/immunology , Cytokines/genetics , Cytokines/immunology , Endocytosis/genetics , Endocytosis/immunology , Filoviridae/genetics , Filoviridae/metabolism , Filoviridae/pathogenicity , Host-Pathogen Interactions/immunology , Interferons/genetics , Interferons/immunology , Orthomyxoviridae/genetics , Orthomyxoviridae/metabolism , Orthomyxoviridae/pathogenicity , Paramyxoviridae/genetics , Paramyxoviridae/metabolism , Paramyxoviridae/pathogenicity , Phosphatidylinositol 3-Kinase/immunology , Pneumovirinae/genetics , Pneumovirinae/metabolism , Pneumovirinae/pathogenicity , Protein Biosynthesis , Proto-Oncogene Proteins c-akt/immunology , RNA Virus Infections/immunology , RNA Virus Infections/virology , Rhabdoviridae/genetics , Rhabdoviridae/metabolism , Rhabdoviridae/pathogenicity , Signal Transduction , Viral Proteins/genetics , Viral Proteins/immunology , Virus Internalization , Virus Replication
4.
Eur J Med Chem ; 204: 112595, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32707357

ABSTRACT

Filoviruses, including Ebolavirus (EBOV), Marburgvirus (MARV) and Cuevavirus, cause hemorrhagic fevers in humans with up to 90% mortality rates. In the 2014-2016 West Africa Ebola epidemic, there are 15,261 laboratory confirmed cases and 11,325 total deaths. The lack of effective vaccines and medicines for the prevention and treatment of filovirus infection in humans stresses the urgency to develop antiviral therapeutics against filovirus-associated diseases. Our previous study identified a histamine receptor antagonist compound CP19 as an entry inhibitor against both EBOV and MARV. The preliminary structure-activity relationship (SAR) studies of CP19 showed that its piperidine, coumarin and linker were related with its antiviral activities. In this study, we performed detailed SAR studies on these groups with synthesized CP19 derivatives. We discovered that 1) the piperidine group could be optimized with heterocycles, 2) the substitution groups of C3 and C4 of coumarin should be relatively large hydrophobic groups and 3) the linker part should be least substituted. Based on the SAR analysis, we synthesized compound 32 as a potent entry inhibitor of EBOV and MARV (IC50 = 0.5 µM for EBOV and 1.5 µM for MARV). The mutation studies of Ebola glycoprotein and molecular docking studies showed that the coumarin and its substituted groups of compound 32 bind to the pocket of Ebola glycoprotein in a similar way to the published entry inhibitor compound 118a. However, the carboxamide group of compound 32 does not have strong interaction with N61 as compound 118a does. The coumarin skeleton structure and the binding model of compound 32 elucidated by this study could be utilized to guide further design and optimization of entry inhibitors targeting the filovirus glycoproteins.


Subject(s)
Coumarins/chemistry , Coumarins/pharmacology , Drug Design , Filoviridae/drug effects , Filoviridae/physiology , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Filoviridae/metabolism , Molecular Targeted Therapy , Piperidines/chemistry , Structure-Activity Relationship
5.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32295912

ABSTRACT

Menglà virus (MLAV), identified in Rousettus bats, is a phylogenetically distinct member of the family Filoviridae Because the filoviruses Ebola virus (EBOV) and Marburg virus (MARV) modulate host innate immunity, MLAV VP35, VP40, and VP24 proteins were compared with their EBOV and MARV homologs for innate immune pathway modulation. In human and Rousettus cells, MLAV VP35 behaved like EBOV and MARV VP35s, inhibiting virus-induced activation of the interferon beta (IFN-ß) promoter and interferon regulatory factor 3 (IRF3) phosphorylation. MLAV VP35 also interacted with PACT, a host protein engaged by EBOV VP35 to inhibit RIG-I signaling. MLAV VP35 also inhibits PKR activation. MLAV VP40 was demonstrated to inhibit type I IFN-induced gene expression in human and bat cells. It blocked STAT1 tyrosine phosphorylation induced either by type I IFN or overexpressed Jak1, paralleling MARV VP40. MLAV VP40 also inhibited virus-induced IFN-ß promoter activation, a property shared by MARV VP40 and EBOV VP24. A Jak kinase inhibitor did not recapitulate this inhibition in the absence of viral proteins. Therefore, inhibition of Jak-STAT signaling is insufficient to explain inhibition of IFN-ß promoter activation. MLAV VP24 did not inhibit IFN-induced gene expression or bind karyopherin α proteins, properties of EBOV VP24. MLAV VP24 differed from MARV VP24 in that it failed to interact with Keap1 or activate an antioxidant response element reporter gene due to the absence of a Keap1-binding motif. These functional observations support a closer relationship of MLAV to MARV than to EBOV but also are consistent with MLAV belonging to a distinct genus.IMPORTANCE EBOV and MARV, members of the family Filoviridae, are highly pathogenic zoonotic viruses that cause severe disease in humans. Both viruses use several mechanisms to modulate the host innate immune response, and these likely contribute to the severity of disease. Here, we demonstrate that MLAV, a filovirus newly discovered in a bat, suppresses antiviral type I interferon responses in both human and bat cells. Inhibitory activities are possessed by MLAV VP35 and VP40, which parallels how MARV blocks IFN responses. However, whereas MARV activates cellular antioxidant responses through an interaction between its VP24 protein and host protein Keap1, MLAV VP24 lacks a Keap1-binding motif and fails to activate this cytoprotective response. These data indicate that MLAV possesses immune-suppressing functions that could facilitate human infection. They also support the placement of MLAV in a different genus than either EBOV or MARV.


Subject(s)
Filoviridae Infections/physiopathology , Filoviridae/genetics , Animals , Chiroptera/immunology , Chiroptera/virology , Ebolavirus , Filoviridae/metabolism , Filoviridae/pathogenicity , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/immunology , Kelch-Like ECH-Associated Protein 1/metabolism , Marburgvirus , NF-E2-Related Factor 2/metabolism , STAT1 Transcription Factor , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
6.
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
7.
PLoS Pathog ; 15(2): e1007564, 2019 02.
Article in English | MEDLINE | ID: mdl-30817809

ABSTRACT

There are a number of vaccine candidates under development against a small number of the most common outbreak filoviruses all employing the virus glycoprotein (GP) as the vaccine immunogen. However, antibodies induced by such GP vaccines are typically autologous and limited to the other members of the same species. In contrast, T-cell vaccines offer a possibility to design a single pan-filovirus vaccine protecting against all known and even likely existing, but as yet unencountered members of the family. Here, we used a cross-filovirus immunogen based on conserved regions of the filovirus nucleoprotein, matrix and polymerase to construct simian adenovirus- and poxvirus MVA-vectored vaccines, and in a proof-of-concept study demonstrated a protection of the BALB/c and C57BL/6J mice against high, lethal challenges with Ebola and Marburg viruses, two distant members of the family, by vaccine-elicited T cells in the absence of GP antibodies.


Subject(s)
Filoviridae/immunology , T-Lymphocytes/immunology , Viral Vaccines/pharmacology , Animals , Antibodies, Neutralizing , Antibodies, Viral , Ebola Vaccines , Ebolavirus/pathogenicity , Female , Filoviridae/metabolism , Filoviridae/pathogenicity , Hemorrhagic Fever, Ebola , Immunity, Cellular/immunology , Male , Marburgvirus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proof of Concept Study , T-Lymphocytes/metabolism
8.
Cell Rep ; 24(10): 2723-2732.e4, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30184505

ABSTRACT

Monoclonal antibodies (mAbs) with pan-ebolavirus cross-reactivity are highly desirable, but development of such mAbs is limited by a lack of a molecular understanding of cross-reactive epitopes. The antibody ADI-15878 was previously identified from a human survivor of Ebola virus Makona variant (EBOV/Mak) infection. This mAb demonstrated potent neutralizing activity against all known ebolaviruses and provided protection in rodent and ferret models against three ebolavirus species. Here, we describe the unliganded crystal structure of ADI-15878 as well as the cryo-EM structures of ADI-15878 in complex with the EBOV/Mak and Bundibugyo virus (BDBV) glycoproteins (GPs). ADI-15878 binds through an induced-fit mechanism by targeting highly conserved residues in the internal fusion loop (IFL), bridging across GP protomers via the heptad repeat 1 (HR1) region. Our structures provide a more complete description of the ebolavirus immunogenic landscape, as well as a molecular basis for how rare but potent antibodies target conserved filoviral fusion machinery.


Subject(s)
Antibodies, Neutralizing/metabolism , Ebolavirus/metabolism , Filoviridae/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Cryoelectron Microscopy , Crystallography , Ebolavirus/genetics , Filoviridae/genetics , Glycoproteins/genetics , Promoter Regions, Genetic/genetics , Protein Structure, Secondary
9.
PLoS One ; 13(8): e0201827, 2018.
Article in English | MEDLINE | ID: mdl-30071116

ABSTRACT

Filoviruses are notorious viral pathogens responsible for high-consequence diseases in humans and non-human primates. Transcription of filovirus mRNA shares several common features with transcription in other non-segmented negative-strand viruses, including differential expression of genes located across the viral genome. Transcriptional patterns of Ebola virus (EBOV) and Marburg virus (MARV) have been previously described using traditional, laborious methods, such as northern blots and in vivo labeling of viral mRNAs. More recently, however, the availability of the next generation sequencing (NGS) technology has offered a more straightforward approach to assess transcriptional patterns. In this report, we analyzed the transcription patterns of four ebolaviruses-EBOV, Sudan (SUDV), Bundibugyo (BDBV), and Reston (RESTV) viruses-in two different cell lines using standard NGS library preparation and sequencing protocols. In agreement with previous reports mainly focused on EBOV and MARV, the remaining filoviruses used in this study also showed a consistent transcription pattern, with only minor variations between the different viruses. We have also analyzed the proportions of the three mRNAs transcribed from the GP gene, which are characteristic of the genus Ebolavirus and encode the glycoprotein (GP), the soluble GP (sGP), and the small soluble GP (ssGP). In addition, we used NGS methodology to analyze the transcription pattern of two previously described recombinant MARV. This analysis allowed us to correct our construction design, and to make an improved version of the original MARV expressing reporter genes.


Subject(s)
Filoviridae Infections/metabolism , Filoviridae/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Transcription, Genetic , Animals , Cell Culture Techniques , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Humans , Liver/metabolism , Liver/virology , Macrophages/metabolism , Macrophages/virology , Temperature
10.
Subcell Biochem ; 88: 73-94, 2018.
Article in English | MEDLINE | ID: mdl-29900493

ABSTRACT

Filoviruses are highly filamentous enveloped animal viruses that can cause severe haemorrhagic fevers. The filovirus ribonucleoprotein forms a highly organized double-layered helical nucleocapsid (NC) containing five different virally encoded proteins. The inner layer consists of NP, the RNA binding protein, complexed with the monopartite linear genome. A distinctive outer layer links individual NP subunits with bridges composed of VP24-VP35 heterodimers, which achieves condensation of the NP-RNA into tight helical coils. There are no vertical connections between the outer helical layers, explaining the flexibility of the NC and its ability to bend into tight curves without breaking the genomic RNA. These properties allow the formation of enveloped virions with varying polymorphisms, including single, linear, continuous, linked, comma-shaped and torroidal forms. Virion length is modular so that just one, or two or more genome copies may be present in each virion, producing polyploid particles. The matrix protein VP40, which drives budding and envelopment, is found in a layer adjacent to the inner cytoplasmic side of viral envelope and is arranged in a 5 nm lattice structure, but its exact symmetry is unclear. There is a constant low density gap between VP40 and the nucleocapsid, so that the latter is held rigidly centred on the long axis of the viral filament. This gap likely contains a region of flexible contacts between VP40 and the NC. The unique morphology of filoviruses may be related to high titre replication, their ease of transmission, and abilities to invade a wide range of host cells and tissues.


Subject(s)
Filoviridae , Genome, Viral/physiology , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , RNA, Viral , RNA-Binding Proteins , Animals , Filoviridae/genetics , Filoviridae/metabolism , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
11.
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
12.
Sci Rep ; 7(1): 15091, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118454

ABSTRACT

Filoviruses are highly virulent pathogens capable of causing severe disease. The glycoproteins of filoviruses are the only virally expressed proteins on the virion surface and are required for receptor binding. As such, they are the main candidate vaccine antigen. Despite their virulence, most filoviruses are not comprehensively characterized, and relatively few commercially produced reagents are available for their study. Here, we describe two methods for production and purification of filovirus glycoproteins in insect and mammalian cell lines. Considerations of expression vector choice, modifications to sequence, troubleshooting of purification method, and glycosylation differences are all important for successful expression of filovirus glycoproteins in cell lines. Given the scarcity of commercially available filovirus glycoproteins, we hope our experiences with possible difficulties in purification of the proteins will facilitate other researchers to produce and purify filovirus glycoproteins rapidly.


Subject(s)
Filoviridae/immunology , Glycoproteins/immunology , Viral Proteins/immunology , Virion/immunology , Animals , Antibodies, Viral/immunology , Filoviridae/metabolism , Filoviridae/pathogenicity , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , Humans , RNA Editing , Sf9 Cells , Spodoptera , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/genetics , Virion/metabolism , Virulence
13.
Curr Top Microbiol Immunol ; 411: 381-417, 2017.
Article in English | MEDLINE | ID: mdl-28795188

ABSTRACT

In this chapter, we describe what is known thus far about the structures and functions of the handful of proteins encoded by filovirus genomes. Amongst the fascinating findings of the last decade is the plurality of functions and structures that these polypeptides can adopt. Many of the encoded proteins can play multiple, distinct roles in the virus life cycle, although the mechanisms by which these functions are determined and controlled remain mostly veiled. Further, some filovirus proteins are multistructural: adopting different oligomeric assemblies and sometimes, different tertiary structures to achieve their separate, and equally essential functions. Structures, and the functions they dictate, are described for components of the nucleocapsid, the matrix, and the surface and secreted glycoproteins.


Subject(s)
Filoviridae/chemistry , Filoviridae/metabolism , Nucleocapsid/chemistry , Nucleocapsid/metabolism
14.
Curr Top Microbiol Immunol ; 411: 353-380, 2017.
Article in English | MEDLINE | ID: mdl-28601948

ABSTRACT

This chapter reviews our current knowledge about the spatiotemporal assembly of filoviral particles. We will follow particles from nucleocapsid entry into the cytoplasm until the nucleocapsids are enveloped at the plasma membrane. We will also highlight the currently open scientific questions surrounding filovirus assembly.


Subject(s)
Filoviridae/chemistry , Filoviridae/metabolism , Virus Assembly , Cell Membrane/virology , Cytoplasm/virology , Nucleocapsid/metabolism
16.
J Virol ; 88(1): 99-109, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24131711

ABSTRACT

Lloviu virus (LLOV), a novel filovirus detected in bats, is phylogenetically distinct from viruses in the genera Ebolavirus and Marburgvirus in the family Filoviridae. While filoviruses are known to cause severe hemorrhagic fever in humans and/or nonhuman primates, LLOV is biologically uncharacterized, since infectious LLOV has never been isolated. To examine the properties of LLOV, we characterized its envelope glycoprotein (GP), which likely plays a key role in viral tropism and pathogenicity. We first found that LLOV GP principally has the same primary structure as the other filovirus GPs. Similar to the other filoviruses, virus-like particles (VLPs) produced by transient expression of LLOV GP, matrix protein, and nucleoprotein in 293T cells had densely arrayed GP spikes on a filamentous particle. Mouse antiserum to LLOV VLP was barely cross-reactive to viruses of the other genera, indicating that LLOV is serologically distinct from the other known filoviruses. For functional study of LLOV GP, we utilized a vesicular stomatitis virus (VSV) pseudotype system and found that LLOV GP requires low endosomal pH and cathepsin L, and that human C-type lectins act as attachment factors for LLOV entry into cells. Interestingly, LLOV GP-pseudotyped VSV infected particular bat cell lines more efficiently than viruses bearing other filovirus GPs. These results suggest that LLOV GP mediates cellular entry in a manner similar to that of the other filoviruses while showing preferential tropism for some bat cells.


Subject(s)
Filoviridae/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cross Reactions , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Filoviridae/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Molecular Sequence Data , Viral Tropism
17.
PLoS One ; 7(10): e44769, 2012.
Article in English | MEDLINE | ID: mdl-23056184

ABSTRACT

We report the rational design and in vivo testing of mosaic proteins for a polyvalent pan-filoviral vaccine using a computational strategy designed for the Human Immunodeficiency Virus type 1 (HIV-1) but also appropriate for Hepatitis C virus (HCV) and potentially other diverse viruses. Mosaics are sets of artificial recombinant proteins that are based on natural proteins. The recombinants are computationally selected using a genetic algorithm to optimize the coverage of potential cytotoxic T lymphocyte (CTL) epitopes. Because evolutionary history differs markedly between HIV-1 and filoviruses, we devised an adapted computational technique that is effective for sparsely sampled taxa; our first significant result is that the mosaic technique is effective in creating high-quality mosaic filovirus proteins. The resulting coverage of potential epitopes across filovirus species is superior to coverage by any natural variants, including current vaccine strains with demonstrated cross-reactivity. The mosaic cocktails are also robust: mosaics substantially outperformed natural strains when computationally tested against poorly sampled species and more variable genes. Furthermore, in a computational comparison of cross-reactive potential a design constructed prior to the Bundibugyo outbreak performed nearly as well against all species as an updated design that included Bundibugyo. These points suggest that the mosaic designs would be more resilient than natural-variant vaccines against future Ebola outbreaks dominated by novel viral variants. We demonstrate in vivo immunogenicity and protection against a heterologous challenge in a mouse model. This design work delineates the likely requirements and limitations on broadly-protective filoviral CTL vaccines.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Filoviridae Infections/immunology , Filoviridae/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Vaccines/immunology , AIDS Vaccines/immunology , Animals , Antibodies, Viral/immunology , Computational Biology/methods , Cross Reactions/immunology , Drug Design , Ebola Vaccines/administration & dosage , Ebola Vaccines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Filoviridae/metabolism , Filoviridae Infections/prevention & control , Filoviridae Infections/virology , Hepacivirus/immunology , Humans , Mice , Mice, Inbred C57BL , Survival Analysis , Viral Hepatitis Vaccines/immunology , Viral Proteins/immunology , Viral Vaccines/administration & dosage
18.
J Virol ; 86(19): 10739-47, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837209

ABSTRACT

Tick-borne Nyamanini virus (NYMV) is the prototypic member of a recently discovered genus in the order Mononegavirales, designated Nyavirus. The NYMV genome codes for six distinct genes. Sequence similarity and structural properties suggest that genes 1, 5, and 6 encode the nucleoprotein (N), the glycoprotein (G), and the viral polymerase (L), respectively. The function of the other viral genes has been unknown to date. We found that the third NYMV gene codes for a protein which, when coexpressed with N and L, can reconstitute viral polymerase activity, suggesting that it represents a polymerase cofactor. The second viral gene codes for a small protein that inhibits viral polymerase activity and further strongly enhances the formation of virus-like particles when coexpressed with gene 4 and the viral glycoprotein G. This suggests that two distinct proteins serve a matrix protein function in NYMV as previously described for members of the family Filoviridae. We further found that NYMV replicates in the nucleus of infected cells like members of the family Bornaviridae. NYMV is a poor inducer of beta interferon, presumably because the viral genome is 5' monophosphorylated and has a protruding 3' terminus as observed for bornaviruses. Taken together, our results demonstrate that NYMV possesses biological properties previously regarded as typical for filoviruses and bornaviruses, respectively.


Subject(s)
Mononegavirales/genetics , Mononegavirales/metabolism , Virus Replication , Animals , Cell Line , Chlorocebus aethiops , Dogs , Filoviridae/metabolism , Genome, Viral , Glycoproteins/chemistry , HEK293 Cells , Humans , Nucleoproteins/chemistry , Phosphorylation , Plasmids/metabolism , Subcellular Fractions/metabolism , Ticks , Vero Cells , Viral Matrix Proteins/metabolism
19.
Immunol Lett ; 141(1): 28-35, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-21911007

ABSTRACT

T cell, immunoglobulin domain and mucin domain-1 (TIM-1) is the nominant member of a small family of related proteins that regulate immune cell activities. TIM-1 was initially characterized in a mouse congenic analysis of Th2 T cell responses and related pathology. Data accumulated to date suggest that TIM-1 regulates effector T cell function, and may play distinct roles in the activities of B cells, invariant NKT cells and epithelial cells. In addition, a variety of ligands for TIM-1 have been proposed. In this review I discuss recent data that have accumulated on the function of TIM-1, propose a model to explain how TIM-1 regulates effector T cell activity through recognition of distinct ligands, and review others functions of this increasingly fascinating protein. Of considerable interest are the novel findings that TIM-1 mediates virus entry and virulence.


Subject(s)
Asthma/immunology , Membrane Glycoproteins , Membrane Proteins , Receptors, Immunologic/metabolism , Receptors, Virus , T-Lymphocytes , Animals , Dendritic Cells/immunology , Filoviridae/metabolism , Hepatitis A Virus Cellular Receptor 1 , Hepatitis A virus/metabolism , Humans , Hygiene Hypothesis , Infections/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/immunology , Protein Isoforms , Receptors, Virus/immunology , Receptors, Virus/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Mol Cell Proteomics ; 9(12): 2690-703, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20702783

ABSTRACT

An assessment of the total protein composition of filovirus (ebolavirus and marburgvirus) virions is currently lacking. In this study, liquid chromatography-linked tandem mass spectrometry of purified ebola and marburg virions was performed to identify associated cellular proteins. Host proteins involved in cell adhesion, cytoskeleton, cell signaling, intracellular trafficking, membrane organization, and chaperones were identified. Significant overlap exists between this data set and proteomic studies of disparate viruses, including HIV-1 and influenza A, generated in multiple cell types. However, the great majority of proteins identified here have not been previously described to be incorporated within filovirus particles. Host proteins identified by liquid chromatography-linked tandem mass spectrometry could lack biological relevance because they represent protein contaminants in the virus preparation, or because they are incorporated within virions by chance. These issues were addressed using siRNA library-mediated gene knockdown (targeting each identified virion-associated host protein), followed by filovirus infection. Knockdown of several host proteins (e.g. HSPA5 and RPL18) significantly interfered with ebolavirus and marburgvirus infection, suggesting specific and relevant virion incorporation. Notably, select siRNAs inhibited ebolavirus, but enhanced marburgvirus infection, suggesting important differences between the two viruses. The proteomic analysis presented here contributes to a greater understanding of filovirus biology and potentially identifies host factors that can be targeted for antiviral drug development.


Subject(s)
Filoviridae/metabolism , Proteomics , RNA Interference , Viral Proteins/metabolism , Virion/metabolism , Amino Acid Sequence , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Gene Knockdown Techniques , Microscopy, Fluorescence , Molecular Sequence Data , Polymerase Chain Reaction , Tandem Mass Spectrometry , Viral Proteins/chemistry , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...