Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Infect Dis ; 218(suppl_5): S666-S671, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30239745

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Glucolípidos/farmacología , Manósidos/uso terapéutico , Animales , Chlorocebus aethiops , Ebolavirus/fisiología , Humanos , Células Vero , Internalización del Virus/efectos de los fármacos
2.
PLoS Pathog ; 10(11): e1004509, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25412102

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas Virales/inmunología , Animales , Antígenos CD/inmunología , Citocinas/inmunología , Células Dendríticas/patología , Células Dendríticas/virología , Cobayas , Células Endoteliales de la Vena Umbilical Humana/patología , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Macrófagos/patología , Macrófagos/virología , Receptor Toll-Like 4/inmunología
3.
J Infect Dis ; 212 Suppl 2: S226-33, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26138826

RESUMEN

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.


Asunto(s)
Ebolavirus/genética , Genes Virales/genética , Fiebre Hemorrágica Ebola/virología , Edición de ARN/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética , Factores de Virulencia/genética , Animales , Línea Celular , Regulación hacia Abajo/genética , Ebolavirus/patogenicidad , Regulación Viral de la Expresión Génica/genética , Cobayas , Glicoproteínas de Membrana/genética , Mutación/genética
4.
J Infect Dis ; 212 Suppl 2: S191-8, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25838269

RESUMEN

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.


Asunto(s)
Ebolavirus/genética , Glicoproteínas/genética , Poliadenilación/genética , Edición de ARN/genética , Proteínas Virales/genética , Animales , Línea Celular , Chlorocebus aethiops , Codón de Terminación/genética , Regulación hacia Abajo/genética , Células HEK293 , Humanos , ARN Mensajero/genética , Células Vero
5.
J Infect Dis ; 212 Suppl 2: S322-8, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26092855

RESUMEN

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.


Asunto(s)
Ebolavirus/metabolismo , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/virología , Glicoproteínas de Membrana/metabolismo , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Chlorocebus aethiops , Ebolavirus/genética , Glicoproteínas de Membrana/genética , Células Vero , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virión/genética , Virión/metabolismo , Virión/patogenicidad , Virulencia/genética , Internalización del Virus , Replicación Viral/genética
6.
J Infect Dis ; 212 Suppl 2: S368-71, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25732811

RESUMEN

The current unprecedented outbreak of Ebola virus (EBOV) disease in West Africa has demonstrated the urgent need for a vaccine. Here, we describe the evaluation of an EBOV vaccine candidate based on Kunjin replicon virus-like particles (KUN VLPs) encoding EBOV glycoprotein with a D637L mutation (GP/D637L) in nonhuman primates. Four African green monkeys (Cercopithecus aethiops) were injected subcutaneously with a dose of 10(9) KUN VLPs per animal twice with an interval of 4 weeks, and animals were challenged 3 weeks later intramuscularly with 600 plaque-forming units of Zaire EBOV. Three animals were completely protected against EBOV challenge, while one vaccinated animal and the control animal died from infection. We suggest that KUN VLPs encoding GP/D637L represent a viable EBOV vaccine candidate.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Replicón/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Virus del Nilo Occidental/inmunología , África Occidental , Animales , Chlorocebus aethiops , Glicoproteínas/inmunología , Inmunización/métodos , Primates , Proteínas Virales/inmunología
7.
Arch Virol ; 159(5): 1229-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24190508

RESUMEN

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.


Asunto(s)
Filoviridae/clasificación , Filoviridae/genética , Virus Reordenados/clasificación , Virus Reordenados/genética , Genoma Viral
8.
J Virol ; 86(19): 10766-75, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22837207

RESUMEN

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.


Asunto(s)
Fosfoproteínas/genética , Fosfoproteínas/fisiología , Proteínas Virales/genética , Proteínas Virales/fisiología , Animales , Cricetinae , Citocinas/metabolismo , Células Endoteliales/citología , Células Endoteliales/virología , Regulación Viral de la Expresión Génica , Células HEK293 , Humanos , Inflamación , Mesocricetus , Microcirculación , Virus Nipah/metabolismo , Proteínas Recombinantes/química , Factores de Tiempo , Venas Umbilicales/citología , Virulencia
9.
Biomolecules ; 13(3)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36979390

RESUMEN

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.


Asunto(s)
Virus Nipah , Dispersión del Ángulo Pequeño , Difracción de Rayos X , Virus Nipah/genética , Virus Nipah/metabolismo , Inmunidad Innata , ARN/metabolismo
10.
J Infect Dis ; 204 Suppl 3: S934-40, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21987772

RESUMEN

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.


Asunto(s)
Ebolavirus/fisiología , Regulación Viral de la Expresión Génica/fisiología , Factores de Transcripción/metabolismo , Proteínas Virales/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Línea Celular , Cricetinae , Factores de Transcripción/genética , Transcripción Genética , Proteínas Virales/genética
11.
J Infect Dis ; 204 Suppl 3: S941-6, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21987773

RESUMEN

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.


Asunto(s)
Ebolavirus/genética , Ebolavirus/fisiología , Genoma Viral/genética , Fiebre Hemorrágica Ebola/virología , Edición de ARN/fisiología , ARN Viral/genética , Adaptación Fisiológica , Animales , Chlorocebus aethiops , Femenino , Regulación Viral de la Expresión Génica/fisiología , Cobayas , Pase Seriado , Células Vero , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/fisiología
12.
J Infect Dis ; 204 Suppl 3: S1011-20, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21987737

RESUMEN

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.


Asunto(s)
Ebolavirus/metabolismo , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/virología , Proteínas Virales/metabolismo , Animales , Línea Celular , Ebolavirus/clasificación , Ebolavirus/genética , Femenino , Regulación Viral de la Expresión Génica/fisiología , Cobayas , Humanos , Hígado/ultraestructura , Macrófagos/virología , Mutación , Virulencia , Replicación Viral
13.
J Infect Dis ; 204 Suppl 3: S892-6, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21987766

RESUMEN

The structural protein VP24 of Ebola virus (EBOV) is a determinant of virulence in rodent models and possesses an interferon antagonist function. In this study, we investigate the role of VP24 in EBOV replication using RNA interference by small interfering RNA to knock down the expression of this protein in virus-infected cells. We reveal that VP24 is required for assembly of viral nucleocapsids and that silencing of VP24 expression prevents the release of EBOV.


Asunto(s)
Ebolavirus/fisiología , Silenciador del Gen , Nucleocápside/fisiología , Proteínas Virales/metabolismo , Ensamble de Virus/fisiología , Replicación Viral/genética , Animales , Chlorocebus aethiops , Ebolavirus/genética , Regulación Viral de la Expresión Génica , Ratones , Conejos , Proteínas Recombinantes , Células Vero , Proteínas Virales/genética , Replicación Viral/fisiología
14.
J Mol Biol ; 434(10): 167551, 2022 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-35317998

RESUMEN

To understand the dynamic interactions between the phosphoprotein (P) and the nucleoprotein (N) within the transcription/replication complex of the Paramyxoviridae and to decipher their roles in regulating viral multiplication, we characterized the structural properties of the C-terminal X domain (PXD) of Nipah (NiV) and Hendra virus (HeV) P protein. In crystals, isolated NiV PXD adopted a two-helix dimeric conformation, which was incompetent for binding its partners, but in complex with the C-terminal intrinsically disordered tail of the N protein (NTAIL), it folded into a canonical 3H bundle conformation. In solution, SEC-MALLS, SAXS and NMR spectroscopy experiments indicated that both NiV and HeV PXD were larger in size than expected for compact proteins of the same molecular mass and were in conformational exchange between a compact three-helix (3H) bundle and partially unfolded conformations, where helix α3 is detached from the other two. Some measurements also provided strong evidence for dimerization of NiV PXD in solution but not for HeV PXD. Ensemble modeling of experimental SAXS data and statistical-dynamical modeling reconciled all these data, yielding a model where NiV and HeV PXD exchanged between different conformations, and where NiV but not HeV PXD formed dimers. Finally, recombinant NiV comprising a chimeric P carrying HeV PXD was rescued and compared with parental NiV. Experiments carried out in cellula demonstrated that the replacement of PXD did not significantly affect the replication dynamics while caused a slight virus attenuation, suggesting a possible role of the dimerization of NiV PXD in viral replication.


Asunto(s)
Virus Hendra , Virus Nipah , Proteínas de la Nucleocápside , Fosfoproteínas , Proteínas Virales , Replicación Viral , Virus Hendra/genética , Virus Hendra/fisiología , Humanos , Virus Nipah/genética , Virus Nipah/fisiología , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Dominios Proteicos , Pliegue de Proteína , Multimerización de Proteína , Dispersión del Ángulo Pequeño , Proteínas Virales/química , Proteínas Virales/genética , Difracción de Rayos X
15.
J Virol ; 84(6): 3004-15, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20071589

RESUMEN

Ebola virus (EBOV) protein VP35 is a double-stranded RNA (dsRNA) binding inhibitor of host interferon (IFN)-alpha/beta responses that also functions as a viral polymerase cofactor. Recent structural studies identified key features, including a central basic patch, required for VP35 dsRNA binding activity. To address the functional significance of these VP35 structural features for EBOV replication and pathogenesis, two point mutations, K319A/R322A, that abrogate VP35 dsRNA binding activity and severely impair its suppression of IFN-alpha/beta production were identified. Solution nuclear magnetic resonance (NMR) spectroscopy and X-ray crystallography reveal minimal structural perturbations in the K319A/R322A VP35 double mutant and suggest that loss of basic charge leads to altered function. Recombinant EBOVs encoding the mutant VP35 exhibit, relative to wild-type VP35 viruses, minimal growth attenuation in IFN-defective Vero cells but severe impairment in IFN-competent cells. In guinea pigs, the VP35 mutant virus revealed a complete loss of virulence. Strikingly, the VP35 mutant virus effectively immunized animals against subsequent wild-type EBOV challenge. These in vivo studies, using recombinant EBOV viruses, combined with the accompanying biochemical and structural analyses directly correlate VP35 dsRNA binding and IFN inhibition functions with viral pathogenesis. Moreover, these studies provide a framework for the development of antivirals targeting this critical EBOV virulence factor.


Asunto(s)
Ebolavirus/genética , Ebolavirus/patogenicidad , Cobayas/virología , Mutación Puntual , ARN Bicatenario/metabolismo , Proteínas Reguladoras y Accesorias Virales , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Ebolavirus/inmunología , Femenino , Humanos , Factores Inmunológicos/genética , Factores Inmunológicos/inmunología , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/genética , Interferón beta/inmunología , Interferones/antagonistas & inhibidores , Interferones/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , ARN Bicatenario/genética , Alineación de Secuencia , Células Vero , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo
16.
J Virol ; 83(16): 7828-41, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19515782

RESUMEN

The Nipah virus (NiV) phosphoprotein (P) gene encodes the C, P, V, and W proteins. P, V, and W, have in common an amino-terminal domain sufficient to bind STAT1, inhibiting its interferon (IFN)-induced tyrosine phosphorylation. P is also essential for RNA-dependent RNA polymerase function. C is encoded by an alternate open reading frame (ORF) within the common amino-terminal domain. Mutations within residues 81 to 113 of P impaired its polymerase cofactor function, as assessed by a minireplicon assay, but these mutants retained STAT1 inhibitory function. Mutations within the residue 114 to 140 region were identified that abrogated interaction with and inhibition of STAT1 by P, V, and W without disrupting P polymerase cofactor function. Recombinant NiVs were then generated. A G121E mutation, which abrogated inhibition of STAT1, was introduced into a C protein knockout background (C(ko)) because the mutation would otherwise also alter the overlapping C ORF. In cell culture, relative to the wild-type virus, the C(ko) mutation proved attenuating but the G121E mutant virus replicated identically to the C(ko) virus. In cells infected with the wild-type and C(ko) viruses, STAT1 was nuclear despite the absence of tyrosine phosphorylation. This latter observation mirrors what has been seen in cells expressing NiV W. In the G121E mutant virus-infected cells, STAT1 was not phosphorylated and was cytoplasmic in the absence of IFN stimulation but became tyrosine phosphorylated and nuclear following IFN addition. These data demonstrate that the gene for NiV P encodes functions that sequester inactive STAT1 in the nucleus, preventing its activation and suggest that the W protein is the dominant inhibitor of STAT1 in NiV-infected cells.


Asunto(s)
Núcleo Celular/metabolismo , Infecciones por Henipavirus/metabolismo , Virus Nipah/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteínas Virales/metabolismo , Proteínas Estructurales Virales/metabolismo , Línea Celular , Núcleo Celular/genética , Infecciones por Henipavirus/virología , Humanos , Mutación , Virus Nipah/genética , Fosfoproteínas/genética , Fosforilación , Unión Proteica , Factor de Transcripción STAT1/genética , Proteínas Virales/genética , Proteínas Estructurales Virales/genética
17.
J Virol ; 83(18): 9596-601, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19587051

RESUMEN

Ebolavirus (EBOV) is the etiological agent of a severe hemorrhagic fever with a high mortality rate. The spike glycoprotein (GP) is believed to be one of the major determinants of virus pathogenicity. In this study, we demonstrated the molecular mechanism responsible for the downregulation of surface markers caused by EBOV GP expression. We showed that expression of mature GP on the plasma membrane results in the masking of cellular surface proteins, including major histocompatibility complex class I. Overexpression of GP also results in the masking of certain antigenic epitopes on GP itself, causing an illusory effect of disappearance from the plasma membrane.


Asunto(s)
Ebolavirus/patogenicidad , Epítopos , Proteínas de la Membrana , Proteínas del Envoltorio Viral/fisiología , Línea Celular , Membrana Celular/virología , Regulación hacia Abajo , Ebolavirus/química , Antígenos de Histocompatibilidad Clase I , Humanos , Transfección , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
18.
Sci Rep ; 7: 41537, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28155869

RESUMEN

Herein we describe production of purified equine IgG obtained from horses immunized with plasmid DNA followed by boosting with Kunjin replicon virus-like particles both encoding a modified Ebola glycoprotein. Administration of the equine IgG over 5 days to cynomolgus macaques infected 24 hours previously with a lethal dose of Ebola virus suppressed viral loads by more than 5 logs and protected animals from mortality. Animals generated their own Ebola glycoprotein-specific IgG responses 9-15 days after infection, with circulating virus undetectable by day 15-17. Such equine IgG may find utility as a post-exposure prophylactic for Ebola infection and provides a low cost, scalable alternative to monoclonal antibodies, with extensive human safety data and WHO-standardized international manufacturing capability available in both high and low income countries.


Asunto(s)
Anticuerpos Antivirales/administración & dosificación , Antígenos Virales/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Inmunoglobulina G/administración & dosificación , Profilaxis Posexposición , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/aislamiento & purificación , Especificidad de Anticuerpos/inmunología , Glicoproteínas/inmunología , Caballos , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Macaca fascicularis
19.
Cell Rep ; 6(6): 1026-1036, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24630992

RESUMEN

Marburg virus (MARV) has a high fatality rate in humans, causing hemorrhagic fever characterized by massive viral replication and dysregulated inflammation. Here, we demonstrate that VP24 of MARV binds Kelch-like ECH-associated protein 1 (Keap1), a negative regulator of nuclear transcription factor erythroid-derived 2 (Nrf2). Binding of VP24 to Keap1 Kelch domain releases Nrf2 from Keap1-mediated inhibition promoting persistent activation of a panoply of cytoprotective genes implicated in cellular responses to oxidative stress and regulation of inflammatory responses. Increased expression of Nrf2-dependent genes was demonstrated both during MARV infection and upon ectopic expression of MARV VP24. We also show that Nrf2-deficient mice can control MARV infection when compared to lethal infection in wild-type animals, indicating that Nrf2 is critical for MARV infection. We conclude that VP24-driven activation of the Nrf2-dependent pathway is likely to contribute to dysregulation of host antiviral inflammatory responses and that it ensures survival of MARV-infected cells despite these responses.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Marburgvirus/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas del Citoesqueleto/metabolismo , Células HEK293 , Humanos , Proteína 1 Asociada A ECH Tipo Kelch , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Transducción de Señal , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA