Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Emerg Infect Dis ; 26(1): 104-113, 2020 01.
Article in English | MEDLINE | ID: mdl-31855143

ABSTRACT

We conducted an in-depth characterization of the Nipah virus (NiV) isolate previously obtained from a Pteropus lylei bat in Cambodia in 2003 (CSUR381). We performed full-genome sequencing and phylogenetic analyses and confirmed CSUR381 is part of the NiV-Malaysia genotype. In vitro studies revealed similar cell permissiveness and replication of CSUR381 (compared with 2 other NiV isolates) in both bat and human cell lines. Sequence alignments indicated conservation of the ephrin-B2 and ephrin-B3 receptor binding sites, the glycosylation site on the G attachment protein, as well as the editing site in phosphoprotein, suggesting production of nonstructural proteins V and W, known to counteract the host innate immunity. In the hamster animal model, CSUR381 induced lethal infections. Altogether, these data suggest that the Cambodia bat-derived NiV isolate has high pathogenic potential and, thus, provide insight for further studies and better risk assessment for future NiV outbreaks in Southeast Asia.


Subject(s)
Chiroptera/virology , Henipavirus Infections/veterinary , Nipah Virus/pathogenicity , Animals , Cambodia , Genome, Viral/genetics , Henipavirus Infections/epidemiology , Henipavirus Infections/virology , Humans , Nipah Virus/genetics , Phylogeny , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Whole Genome Sequencing
2.
J Infect Dis ; 218(suppl_5): S666-S671, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30239745

ABSTRACT

The West African outbreak of Ebola virus (EBOV) infection during 2013-2016 highlighted the need for development of field-applicable therapeutic drugs for this infection. Here we report that mannoside glycolipid conjugates (MGCs) consisting of a trimannose head and a lipophilic chain assembled by a linker inhibit EBOV infection not only of human monocyte-derived dendritic cells and macrophages, but also of a number of susceptible cells. Analysis of the mode of action leads us to conclude that MGCs act directly on cells, notably by preventing virus endocytosis.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Glycolipids/pharmacology , Mannosides/therapeutic use , Animals , Chlorocebus aethiops , Ebolavirus/physiology , Humans , Vero Cells , Virus Internalization/drug effects
3.
PLoS Pathog ; 10(11): e1004509, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25412102

ABSTRACT

During Ebola virus (EBOV) infection a significant amount of surface glycoprotein GP is shed from infected cells in a soluble form due to cleavage by cellular metalloprotease TACE. Shed GP and non-structural secreted glycoprotein sGP, both expressed from the same GP gene, have been detected in the blood of human patients and experimentally infected animals. In this study we demonstrate that shed GP could play a particular role during EBOV infection. In effect it binds and activates non-infected dendritic cells and macrophages inducing the secretion of pro- and anti-inflammatory cytokines (TNFα, IL1ß, IL6, IL8, IL12p40, and IL1-RA, IL10). Activation of these cells by shed GP correlates with the increase in surface expression of co-stimulatory molecules CD40, CD80, CD83 and CD86. Contrary to shed GP, secreted sGP activates neither DC nor macrophages while it could bind DCs. In this study, we show that shed GP activity is likely mediated through cellular toll-like receptor 4 (TLR4) and is dependent on GP glycosylation. Treatment of cells with anti-TLR4 antibody completely abolishes shed GP-induced activation of cells. We also demonstrate that shed GP activity is negated upon addition of mannose-binding sera lectin MBL, a molecule known to interact with sugar arrays present on the surface of different microorganisms. Furthermore, we highlight the ability of shed GP to affect endothelial cell function both directly and indirectly, demonstrating the interplay between shed GP, systemic cytokine release and increased vascular permeability. In conclusion, shed GP released from virus-infected cells could activate non-infected DCs and macrophages causing the massive release of pro- and anti-inflammatory cytokines and effect vascular permeability. These activities could be at the heart of the excessive and dysregulated inflammatory host reactions to infection and thus contribute to high virus pathogenicity.


Subject(s)
Dendritic Cells/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Human Umbilical Vein Endothelial Cells/immunology , Macrophages/immunology , Membrane Glycoproteins/immunology , Viral Proteins/immunology , Animals , Antigens, CD/immunology , Cytokines/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Guinea Pigs , Human Umbilical Vein Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/virology , Humans , Macrophages/pathology , Macrophages/virology , Toll-Like Receptor 4/immunology
4.
J Infect Dis ; 212 Suppl 2: S372-8, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26232760

ABSTRACT

Ebola virus is the etiological agent of a severe hemorrhagic fever with a high mortality rate. As the only protein exposed on the surface of viral particles, the spike glycoprotein GP is the unique target for neutralizing monoclonal antibodies. In this study, we demonstrate the strong neutralization capacity of the monoclonal antibody #3327 and characterize its activity. GP residues that are required for recognition and neutralization were found to be located both in the internal fusion loop and in the receptor-binding domain. Analysis of Ebola virus entry in the presence of #3327 allows us to hypothesize that this antibody binds to the virus particle before internalization and endosomal processing of GP and likely prevents the final viral fusion step. Importantly, #3327 is able to block entry of virions bearing GP that contain the Q508 escape mutation common to a number of virus-neutralizing antibodies, and therefore provides future perspectives for treatment strategies against Ebola virus infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Animals , Cell Line , Chlorocebus aethiops , Glycoproteins/immunology , HEK293 Cells , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Humans , Protein Binding/immunology , Vero Cells , Virion/immunology , Virus Internalization
5.
J Infect Dis ; 212 Suppl 2: S199-203, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25941332

ABSTRACT

Ebola virus (EBOV) is responsible for a severe fever with a high mortality rate. The diverse nature of the attachment of the virus to the cell surface, the initial step of virus entry, raises questions concerning the kinetics of the virus internalization process. We investigated EBOV entry kinetics using the activity of a particular monoclonal antibody that neutralizes virus infectivity. We demonstrate that inoculation of cells with EBOV results in an asynchronous entry process, as revealed by the ability of the virus to remain in a cell-bound state for an extended period of time before it is internalized.


Subject(s)
Ebolavirus/physiology , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Humans , Kinetics , Vero Cells , Virus Internalization
6.
J Infect Dis ; 212 Suppl 2: S226-33, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26138826

ABSTRACT

Synthesis of the surface glycoprotein GP of Ebola virus (EBOV) is dependent on transcriptional RNA editing, whereas direct expression of the GP gene results in synthesis of nonstructural secreted glycoprotein sGP. In this study, we investigate the role of RNA editing in the pathogenicity of EBOV using a guinea pig model and recombinant guinea pig-adapted EBOV containing mutations at the editing site, allowing expression of surface GP without the need for RNA editing, and also preventing synthesis of sGP. We demonstrate that the elimination of the editing site leads to EBOV attenuation in vivo, explained by lower virus spread caused by the higher virus cytotoxicity and, most likely, by an increased ability of the host defense systems to recognize and eliminate virus-infected cells. We also demonstrate that expression of sGP does not affect pathogenicity of EBOV in guinea pigs. In conclusion, data obtained indicate that downregulation of the level of surface GP expression through a mechanism of GP gene RNA editing plays an important role in the high pathogenicity of EBOV.


Subject(s)
Ebolavirus/genetics , Genes, Viral/genetics , Hemorrhagic Fever, Ebola/virology , RNA Editing/genetics , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Virulence Factors/genetics , Animals , Cell Line , Down-Regulation/genetics , Ebolavirus/pathogenicity , Gene Expression Regulation, Viral/genetics , Guinea Pigs , Membrane Glycoproteins/genetics , Mutation/genetics
7.
J Infect Dis ; 212 Suppl 2: S191-8, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25838269

ABSTRACT

Synthesis of Ebola virus (EBOV) surface glycoprotein (GP) is dependent on transcriptional RNA editing. Northern blot analysis of EBOV-infected cells using GP-gene-specific probes reveals that, in addition to full-length GP messenger RNAs (mRNAs), a shorter RNA is also synthesized, representing >40% of the total amount of GP mRNA. Sequence analysis demonstrates that this RNA is a truncated version of the full-length GP mRNA that is polyadenylated at the editing site and thus lacks a stop codon. An absence of detectable levels of protein synthesis in cellulo is consistent with the existence of tight regulation of the translation of such mRNA. However, nonstop GP mRNA was shown to be only slightly less stable than the same mRNA containing a stop codon, against the general belief in nonstop decay mechanisms aimed at detecting and destroying mRNAs lacking a stop codon. In conclusion, we demonstrate that the editing site indeed serves as a cryptic transcription termination/polyadenylation site, which rarely also functions to edit GP mRNA for expression of surface GP. This new data suggest that the downregulation of surface GP expression is even more dramatic than previously thought, reinforcing the importance of the GP gene editing site for EBOV replication and pathogenicity.


Subject(s)
Ebolavirus/genetics , Glycoproteins/genetics , Polyadenylation/genetics , RNA Editing/genetics , Viral Proteins/genetics , Animals , Cell Line , Chlorocebus aethiops , Codon, Terminator/genetics , Down-Regulation/genetics , HEK293 Cells , Humans , RNA, Messenger/genetics , Vero Cells
8.
J Infect Dis ; 212 Suppl 2: S322-8, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26092855

ABSTRACT

The surface glycoprotein (GP) is responsible for Ebola virus (EBOV) attachment and membrane fusion during virus entry. Surface expression of highly glycosylated GP causes marked cytotoxicity via masking of a wide range of cellular surface molecules, including integrins. Considerable amounts of surface GP are shed from virus-infected cells in a soluble truncated form by tumor necrosis factor α-converting enzyme. In this study, the role of GP shedding was investigated using a reverse genetics approach by comparing recombinant viruses possessing amino acid substitutions at the GP shedding site. Virus with an L635V substitution showed a substantial decrease in shedding, whereas a D637V substitution resulted in a striking increase in the release of shed GP. Variations in shedding efficacy correlated with observed differences in the amounts of shed GP in the medium, GP present in virus-infected cells, and GP present on virions. An increase in shedding appeared to be associated with a reduction in viral cytotoxicity, and, vice versa, the virus that shed less was more cytotoxic. An increase in shedding also resulted in a reduction in viral infectivity, whereas a decrease in shedding efficacy enhanced viral growth characteristics in vitro. Differences in shedding efficacy and, as a result, differences in the amount of mature GP available for incorporation into budding virions did not equate to differences in overall release of viral particles. Likewise, data suggest that the resulting differences in the amount of mature GP on the cell surface led to variations in the GP content of released particles and, as a consequence, in infectivity. In conclusion, fine-tuning of the levels of EBOV GP expressed at the surface of virus-infected cells via GP shedding plays an important role in EBOV replication by orchestrating the balance between optimal virion GP content and cytotoxicity caused by GP.


Subject(s)
Ebolavirus/metabolism , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Membrane Glycoproteins/metabolism , Amino Acid Substitution/genetics , Animals , Cell Line , Chlorocebus aethiops , Ebolavirus/genetics , Membrane Glycoproteins/genetics , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/genetics , Virion/metabolism , Virion/pathogenicity , Virulence/genetics , Virus Internalization , Virus Replication/genetics
9.
Arch Virol ; 159(4): 821-30, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24122154

ABSTRACT

The International Committee on Taxonomy of Viruses (ICTV) Filoviridae Study Group prepares proposals on the classification and nomenclature of filoviruses to reflect current knowledge or to correct disagreements with the International Code of Virus Classification and Nomenclature (ICVCN). In recent years, filovirus taxonomy has been corrected and updated, but parts of it remain controversial, and several topics remain to be debated. This article summarizes the decisions and discussion of the currently acting ICTV Filoviridae Study Group since its inauguration in January 2012.


Subject(s)
Classification/methods , Filoviridae/classification , Terminology as Topic , Humans
10.
Arch Virol ; 159(5): 1229-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24190508

ABSTRACT

Specific alterations (mutations, deletions, insertions) of virus genomes are crucial for the functional characterization of their regulatory elements and their expression products, as well as a prerequisite for the creation of attenuated viruses that could serve as vaccine candidates. Virus genome tailoring can be performed either by using traditionally cloned genomes as starting materials, followed by site-directed mutagenesis, or by de novo synthesis of modified virus genomes or parts thereof. A systematic nomenclature for such recombinant viruses is necessary to set them apart from wild-type and laboratory-adapted viruses, and to improve communication and collaborations among researchers who may want to use recombinant viruses or create novel viruses based on them. A large group of filovirus experts has recently proposed nomenclatures for natural and laboratory animal-adapted filoviruses that aim to simplify the retrieval of sequence data from electronic databases. Here, this work is extended to include nomenclature for filoviruses obtained in the laboratory via reverse genetics systems. The previously developed template for natural filovirus genetic variant naming, (/)///-, is retained, but we propose to adapt the type of information added to each field for cDNA clone-derived filoviruses. For instance, the full-length designation of an Ebola virus Kikwit variant rescued from a plasmid developed at the US Centers for Disease Control and Prevention could be akin to "Ebola virus H.sapiens-rec/COD/1995/Kikwit-abc1" (with the suffix "rec" identifying the recombinant nature of the virus and "abc1" being a placeholder for any meaningful isolate designator). Such a full-length designation should be used in databases and the methods section of publications. Shortened designations (such as "EBOV H.sap/COD/95/Kik-abc1") and abbreviations (such as "EBOV/Kik-abc1") could be used in the remainder of the text, depending on how critical it is to convey information contained in the full-length name. "EBOV" would suffice if only one EBOV strain/variant/isolate is addressed.


Subject(s)
Filoviridae/classification , Filoviridae/genetics , Reassortant Viruses/classification , Reassortant Viruses/genetics , Genome, Viral
11.
J Virol ; 86(19): 10766-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837207

ABSTRACT

Nipah virus (NiV) is a highly pathogenic, negative-strand RNA paramyxovirus that has recently emerged from flying foxes to cause serious human disease. We have analyzed the role of the nonstructural NiV C protein in viral immunopathogenesis using recombinant virus lacking the expression of NiV C (NiVΔC). While wild-type NiV was highly pathogenic in the hamster animal model, NiVΔC was strongly attenuated. Replication of NiVΔC was followed by the production of NiV-specific antibodies and associated with higher recruitment of inflammatory cells and less intensive histopathological lesions in different organs than in wild-type-NiV-infected animals. To analyze the molecular basis of NiVΔC attenuation, we studied early changes in gene expression in infected primary human endothelial cells, a major cellular target of NiV infection. The transcriptomic approach revealed the striking difference between wild-type and mutant NiV in the expression of genes involved in immunity, with the particularly interesting differential patterns of proinflammatory cytokines. Compared to wild-type virus, NiVΔC induced increased expression of interleukin 1 beta (IL-1ß), IL-8, CXCL2, CXCL3, CXCL6, CCL20, and beta interferon. Furthermore, the expression of NiV C in stably transfected cells decreased the production of the same panel of cytokines, revealing a role of the C protein in the regulation of cytokine balance. Together, these results suggest that NiV C regulates expression of proinflammatory cytokines, therefore providing a signal responsible for the coordination of leukocyte recruitment and the chemokine-induced immune response and controlling the lethal outcome of the infection.


Subject(s)
Phosphoproteins/genetics , Phosphoproteins/physiology , Viral Proteins/genetics , Viral Proteins/physiology , Animals , Cricetinae , Cytokines/metabolism , Endothelial Cells/cytology , Endothelial Cells/virology , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Inflammation , Mesocricetus , Microcirculation , Nipah Virus/metabolism , Recombinant Proteins/chemistry , Time Factors , Umbilical Veins/cytology , Virulence
12.
Biomolecules ; 13(3)2023 03 01.
Article in English | MEDLINE | ID: mdl-36979390

ABSTRACT

The protein C is a small viral protein encoded in an overlapping frame of the P gene in the subfamily Orthoparamyxovirinae. This protein, expressed by alternative translation initiation, is a virulence factor that regulates viral transcription, replication, and production of defective interfering RNA, interferes with the host-cell innate immunity systems and supports the assembly of viral particles and budding. We expressed and purified full-length and an N-terminally truncated C protein from Tupaia paramyxovirus (TupV) C protein (genus Narmovirus). We solved the crystal structure of the C-terminal part of TupV C protein at a resolution of 2.4 Å and found that it is structurally similar to Sendai virus C protein, suggesting that despite undetectable sequence conservation, these proteins are homologous. We characterized both truncated and full-length proteins by SEC-MALLS and SEC-SAXS and described their solution structures by ensemble models. We established a mini-replicon assay for the related Nipah virus (NiV) and showed that TupV C inhibited the expression of NiV minigenome in a concentration-dependent manner as efficiently as the NiV C protein. A previous study found that the Orthoparamyxovirinae C proteins form two clusters without detectable sequence similarity, raising the question of whether they were homologous or instead had originated independently. Since TupV C and SeV C are representatives of these two clusters, our discovery that they have a similar structure indicates that all Orthoparamyxovirine C proteins are homologous. Our results also imply that, strikingly, a STAT1-binding site is encoded by exactly the same RNA region of the P/C gene across Paramyxovirinae, but in different reading frames (P or C), depending on which cluster they belong to.


Subject(s)
Nipah Virus , Scattering, Small Angle , X-Ray Diffraction , Nipah Virus/genetics , Nipah Virus/metabolism , Immunity, Innate , RNA/metabolism
13.
J Virol ; 85(17): 8502-13, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21697477

ABSTRACT

With the exception of Reston and Lloviu viruses, filoviruses (marburgviruses, ebolaviruses, and "cuevaviruses") cause severe viral hemorrhagic fevers in humans. Filoviruses use a class I fusion protein, GP(1,2), to bind to an unknown, but shared, cell surface receptor to initiate virus-cell fusion. In addition to GP(1,2), ebolaviruses and cuevaviruses, but not marburgviruses, express two secreted glycoproteins, soluble GP (sGP) and small soluble GP (ssGP). All three glycoproteins have identical N termini that include the receptor-binding region (RBR) but differ in their C termini. We evaluated the effect of the secreted ebolavirus glycoproteins on marburgvirus and ebolavirus cell entry, using Fc-tagged recombinant proteins. Neither sGP-Fc nor ssGP-Fc bound to filovirus-permissive cells or inhibited GP(1,2)-mediated cell entry of pseudotyped retroviruses. Surprisingly, several Fc-tagged Δ-peptides, which are small C-terminal cleavage products of sGP secreted by ebolavirus-infected cells, inhibited entry of retroviruses pseudotyped with Marburg virus GP(1,2), as well as Marburg virus and Ebola virus infection in a dose-dependent manner and at low molarity despite absence of sequence similarity to filovirus RBRs. Fc-tagged Δ-peptides from three ebolaviruses (Ebola virus, Sudan virus, and Taï Forest virus) inhibited GP(1,2)-mediated entry and infection of viruses comparably to or better than the Fc-tagged RBRs, whereas the Δ-peptide-Fc of an ebolavirus nonpathogenic for humans (Reston virus) and that of an ebolavirus with lower lethality for humans (Bundibugyo virus) had little effect. These data indicate that Δ-peptides are functional components of ebolavirus proteomes. They join cathepsins and integrins as novel modulators of filovirus cell entry, might play important roles in pathogenesis, and could be exploited for the synthesis of powerful new antivirals.


Subject(s)
Antiviral Agents/metabolism , Ebolavirus/drug effects , Immunoglobulin Fc Fragments/metabolism , Marburgvirus/drug effects , Viral Proteins/metabolism , Virus Internalization/drug effects , Animals , Biological Products/metabolism , Cell Line , Ebolavirus/physiology , Humans , Immunoglobulin Fc Fragments/genetics , Marburgvirus/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Proteins/genetics
14.
Nat Med ; 11(7): 786-90, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15937495

ABSTRACT

Vaccines and therapies are urgently needed to address public health needs stemming from emerging pathogens and biological threat agents such as the filoviruses Ebola virus (EBOV) and Marburg virus (MARV). Here, we developed replication-competent vaccines against EBOV and MARV based on attenuated recombinant vesicular stomatitis virus vectors expressing either the EBOV glycoprotein or MARV glycoprotein. A single intramuscular injection of the EBOV or MARV vaccine elicited completely protective immune responses in nonhuman primates against lethal EBOV or MARV challenges. Notably, vaccine vector shedding was not detectable in the monkeys and none of the animals developed fever or other symptoms of illness associated with vaccination. The EBOV vaccine induced humoral and apparent cellular immune responses in all vaccinated monkeys, whereas the MARV vaccine induced a stronger humoral than cellular immune response. No evidence of EBOV or MARV replication was detected in any of the protected animals after challenge. Our data suggest that these vaccine candidates are safe and highly efficacious in a relevant animal model.


Subject(s)
Ebolavirus/immunology , Marburgvirus/immunology , Vaccines, Attenuated/immunology , Vaccines, Combined/immunology , Viral Vaccines/immunology , Animals , Antibody Formation , Cross Reactions , Ebola Vaccines/immunology , Ebola Vaccines/pharmacology , Primates , Vaccines, Attenuated/genetics , Vaccines, Attenuated/pharmacology , Vaccines, Combined/genetics , Vaccines, Combined/pharmacology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology , Vesicular stomatitis Indiana virus/genetics , Viral Vaccines/genetics , Viral Vaccines/pharmacology , Viremia/immunology , Viremia/virology , Virus Replication
15.
J Infect Dis ; 204 Suppl 3: S884-91, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987765

ABSTRACT

The matrix protein VP40 is essential for Ebola virus (EBOV) and Marburg virus assembly and budding at the plasma membrane. In this study we have investigated the effect of single amino acid substitutions in a conserved proline-rich region of the EBOV VP40 located in the carboxy-terminal part of the protein. We demonstrate that substitutions within this region result in an alteration of intracellular VP40 localization and also cause a reduction or a complete block of virus-like particle budding, a benchmark of VP40 function. Furthermore, some mutated VP40s revealed an enhanced binding with cellular Sec24C, a part of the coat protein complex II (COPII) vesicular transport system. Analysis of the 3-dimensional structure of VP40 revealed the spatial proximity of the proline-rich region and an earlier identified site of interaction with Sec24C, thus allowing us to hypothesize that the altered intracellular localization of the VP40 mutants is a consequence of defects in their interaction with COPII-mediated vesicular transport.


Subject(s)
Cell Membrane/metabolism , Ebolavirus/metabolism , Proline/chemistry , Viral Matrix Proteins/metabolism , Virus Release/physiology , Animals , Chlorocebus aethiops , HEK293 Cells , Humans , Models, Molecular , Nocodazole/pharmacology , Protein Conformation , Protein Transport , Tubulin Modulators/pharmacology , Vero Cells , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
16.
J Infect Dis ; 204 Suppl 3: S934-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987772

ABSTRACT

Ebola virus (EBOV) transcription is dependent on the phosphoprotein VP30, a component of the viral nucleocapsid. VP30 is phosphorylated at 2 serine residue clusters located at the N-terminal part of the protein. In this report, we have investigated the role of VP30 phosphorylation in EBOV replication using a reverse genetics approach. In effect, recombinant EBOVs with the VP30 serine clusters substituted either by nonphosphorylatable alanines or phosphorylation-mimicking aspartates were generated and characterized. We show that in comparison to the wild-type EBOV the mutated viruses possess reduced infectivity. This difference is explained by alterations in the balance between the transcription and replication processes and appear to be associated with the state of VP30 phosphorylation. Here we propose a model in which dynamic phosphorylation of VP30 is an important mechanism to regulate the EBOV replication cycle.


Subject(s)
Ebolavirus/physiology , Gene Expression Regulation, Viral/physiology , Transcription Factors/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Cricetinae , Transcription Factors/genetics , Transcription, Genetic , Viral Proteins/genetics
17.
J Infect Dis ; 204 Suppl 3: S941-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987773

ABSTRACT

Synthesis of the structural, surface glycoprotein (GP) of Ebola virus (EBOV) is dependent on transcriptional RNA editing phenomenon. Editing results in the insertion of an extra adenosine by viral polymerase at the editing site (7 consecutive template uridines) during transcription of GP gene of the wild-type virus (EBOV/7U). In this study, we demonstrate that passage of EBOV/7U in Vero E6 cells results in the appearance and rapid accumulation of a variant (EBOV/8U) containing an additional uridine at the editing site in the viral genome. EBOV/8U outgrows and eventually replaces the wild-type EBOV during 4-5 passages. On the contrary, infection of guinea pigs with EBOV/8U leads to the appearance and rapid predominance by EBOV/7U. These rapid conversions suggest that editing of the genomic RNA occurs at a higher frequency than previously thought. In addition, it indicates that the EBOV/7U phenotype has a selective advantage that is linked to controlled expression of GP and/or expression of secreted sGP, the primary gene product for wild-type EBOV. This study demonstrates the potential for insertion and deletion of uridines in the editing site of the EBOV genomic RNA, depending on environmental constraints.


Subject(s)
Ebolavirus/genetics , Ebolavirus/physiology , Genome, Viral/genetics , Hemorrhagic Fever, Ebola/virology , RNA Editing/physiology , RNA, Viral/genetics , Adaptation, Physiological , Animals , Chlorocebus aethiops , Female , Gene Expression Regulation, Viral/physiology , Guinea Pigs , Serial Passage , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/physiology
18.
J Infect Dis ; 204 Suppl 3: S973-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987778

ABSTRACT

We examined the ability of the Ebola virus to elicit an antiviral response from plasmacytoid dendritic cells (pDCs). Exposure of pDCs to Ebola virus did not result in significantly higher levels of interferon-α production than the levels in mock-infected cells. After inoculation with Ebola virus under the same conditions, conventional dendritic cells expressed viral proteins whereas pDCs did not, suggesting that the latter cells were not infected. Assessment of the entry of Ebola virus-like particles into pDCs revealed that pDCs are highly impaired for viral entry in comparison with conventional dendritic cells. These observations identify a novel means by which Ebola virus can avoid triggering an antiviral response.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Ebolavirus/physiology , Interferons/metabolism , Gene Expression Regulation, Viral/physiology , Humans , Viral Proteins/metabolism , Virus Internalization
19.
J Infect Dis ; 204 Suppl 3: S1011-20, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987737

ABSTRACT

In sharp contrast to human and nonhuman primates, guinea pigs and some other mammals resist Ebola virus (EBOV) replication and do not develop illness upon virus inoculation. However, serial passaging of EBOV in guinea pigs results in a selection of variants with high pathogenicity. In this report, using a reverse genetics approach, we demonstrate that this dramatic increase in EBOV pathogenicity is associated with amino acid substitutions in the structural protein VP24. We show that although replication of recombinant EBOV carrying wild-type VP24 is impaired in primary peritoneal guinea pig macrophages and in the liver of infected animals, the substitutions in VP24 allow EBOV to replicate in guinea pig macrophages and spread in the liver of infected animals. Furthermore, we demonstrate that both VP24/wild type and the guinea pig-adapted VP24/8mc are similar in their ability to block expression of interferon-induced host genes, suggesting that the increase in EBOV virulence for guinea pigs is not associated with VP24 interferon antagonist function. This study sheds light on the mechanism of resistance to EBOV infection and highlights the critical role of VP24 in EBOV pathogenesis.


Subject(s)
Ebolavirus/metabolism , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Viral Proteins/metabolism , Animals , Cell Line , Ebolavirus/classification , Ebolavirus/genetics , Female , Gene Expression Regulation, Viral/physiology , Guinea Pigs , Humans , Liver/ultrastructure , Macrophages/virology , Mutation , Virulence , Virus Replication
20.
J Infect Dis ; 204 Suppl 3: S833-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987759

ABSTRACT

The Ebola virus matrix protein VP40 plays an essential role in virus assembly and budding. In this study we reveal that transient VP40 expression results in the release into the culture medium of substantial amounts of soluble monomeric VP40 in addition to the release of virus-like particles containing an oligomeric form of this protein as previously described. We show that VP40 secretion is endoplasmic reticulum/Golgi-independent and is not associated with cell death. Soluble VP40 was observed during Ebola virus infection of cells and was also found in the serum of virus-infected animals albeit in lower amounts. Unconventional secretion of VP40 may therefore play a role in Ebola virus pathogenicity.


Subject(s)
Ebolavirus/metabolism , Viral Matrix Proteins/metabolism , Animals , Chlorocebus aethiops , Culture Media , Ebolavirus/pathogenicity , Endoplasmic Reticulum , Gene Expression Regulation, Viral/physiology , Golgi Apparatus , HEK293 Cells , Humans , Vero Cells , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL