Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 13(3)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36979390

RESUMO

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.


Assuntos
Vírus Nipah , Espalhamento a Baixo Ângulo , Difração de Raios X , Vírus Nipah/genética , Vírus Nipah/metabolismo , Imunidade Inata , RNA/metabolismo
2.
J Mol Biol ; 434(10): 167551, 2022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35317998

RESUMO

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.


Assuntos
Vírus Hendra , Vírus Nipah , Proteínas do Nucleocapsídeo , Fosfoproteínas , Proteínas Virais , Replicação Viral , Vírus Hendra/genética , Vírus Hendra/fisiologia , Humanos , Vírus Nipah/genética , Vírus Nipah/fisiologia , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Domínios Proteicos , Dobramento de Proteína , Multimerização Proteica , Espalhamento a Baixo Ângulo , Proteínas Virais/química , Proteínas Virais/genética , Difração de Raios X
3.
Med Sci (Paris) ; 37(2): 135-140, 2021 Feb.
Artigo em Francês | MEDLINE | ID: mdl-33591256

RESUMO

The Crimean-Congo hemorrhagic fever virus (CCHFV) is the etiological agent of a severe hemorrhagic fever affecting Africa, Asia and southern Europe. Climate changes of recent decades have recently led to a rise in the distribution of this virus. Still few scientific data are available on the biology of its vector, the tick, or its own biology, but the proven presence of human infections observed in Spain and animals with positive serology in Corsica should focus our attention on this pathogen. This review takes stock of the epidemiologic evolution of CCHF in Europe, notably in France.


TITLE: La fièvre hémorragique de Crimée-Congo, une future problématique de santé en France ? ABSTRACT: Le virus de la fièvre hémorragique de Crimée-Congo (CCHFV) est l'agent étiologique d'une fièvre hémorragique grave affectant l'Afrique, l'Asie et le sud de l'Europe. Les modifications climatiques de ces dernières décennies induisent depuis peu une remontée de l'aire de distribution de ce virus. Encore peu de données scientifiques sont disponibles sur les interactions avec son vecteur, la tique, ou sur sa biologie propre. Cependant, la présence avérée d'infections humaines en Espagne et des sérologies positives dans le cheptel corse pourraient bien concentrer l'attention sur ce pathogène. Cette revue fait le point sur l'évolution des connaissances éco-épidémiologiques de ce virus, notamment en Europe et plus particulièrement en France.


Assuntos
Febre Hemorrágica da Crimeia/epidemiologia , Animais , Europa (Continente)/epidemiologia , França/epidemiologia , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Febre Hemorrágica da Crimeia/virologia , Humanos , Estudos Soroepidemiológicos
4.
Elife ; 92020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33349334

RESUMO

The intricate lattice of Gn and Gc glycoprotein spike complexes on the hantavirus envelope facilitates host-cell entry and is the primary target of the neutralizing antibody-mediated immune response. Through study of a neutralizing monoclonal antibody termed mAb P-4G2, which neutralizes the zoonotic pathogen Puumala virus (PUUV), we provide a molecular-level basis for antibody-mediated targeting of the hantaviral glycoprotein lattice. Crystallographic analysis demonstrates that P-4G2 binds to a multi-domain site on PUUV Gc and may preclude fusogenic rearrangements of the glycoprotein that are required for host-cell entry. Furthermore, cryo-electron microscopy of PUUV-like particles in the presence of P-4G2 reveals a lattice-independent configuration of the Gc, demonstrating that P-4G2 perturbs the (Gn-Gc)4 lattice. This work provides a structure-based blueprint for rationalizing antibody-mediated targeting of hantaviruses.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Virus Puumala/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Monoclonais/imunologia , Arvicolinae , Células HEK293 , Humanos
5.
Med Sci (Paris) ; 36(11): 1027-1033, 2020 Nov.
Artigo em Francês | MEDLINE | ID: mdl-33151849

RESUMO

In the recent years, Ebola virus has been responsible for several major epidemics. Research efforts have allowed the development and evaluation in the field of several vaccine candidates. At present, two of them are already approved and used in the fight against the virus in the Democratic Republic of Congo. This review aims to describe the different candidates, the clinical trials that have been conducted as well as the first results in the field.


TITLE: Ebola, des premiers vaccins disponibles. ABSTRACT: Ces dernières années, le virus Ebola a été responsable d'épidémies de grande ampleur. Les efforts de recherche ont permis la mise au point et l'évaluation sur le terrain de plusieurs candidats vaccins. À l'heure actuelle, deux sont déjà homologués et utilisés dans la lutte contre le virus en République démocratique du Congo. Cette revue se propose de faire le point sur les différents candidats vaccins, les essais cliniques qui ont été menés et les premiers résultats de terrain.


Assuntos
Vacinas contra Ebola/uso terapêutico , Ebolavirus/imunologia , Doença pelo Vírus Ebola/terapia , Surtos de Doenças , Epidemias , Necessidades e Demandas de Serviços de Saúde , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/imunologia , Humanos , Vacinação em Massa/organização & administração , Vacinação em Massa/estatística & dados numéricos , Fatores Socioeconômicos
6.
Biophys J ; 118(10): 2470-2488, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32348724

RESUMO

The structural characterization of modular proteins containing long intrinsically disordered regions intercalated with folded domains is complicated by their conformational diversity and flexibility and requires the integration of multiple experimental approaches. Nipah virus (NiV) phosphoprotein, an essential component of the viral RNA transcription/replication machine and a component of the viral arsenal that hijacks cellular components and counteracts host immune responses, is a prototypical model for such modular proteins. Curiously, the phosphoprotein of NiV is significantly longer than the corresponding protein of other paramyxoviruses. Here, we combine multiple biophysical methods, including x-ray crystallography, NMR spectroscopy, and small angle x-ray scattering, to characterize the structure of this protein and provide an atomistic representation of the full-length protein in the form of a conformational ensemble. We show that full-length NiV phosphoprotein is tetrameric, and we solve the crystal structure of its tetramerization domain. Using NMR spectroscopy and small angle x-ray scattering, we show that the long N-terminal intrinsically disordered region and the linker connecting the tetramerization domain to the C-terminal X domain exchange between multiple conformations while containing short regions of residual secondary structure. Some of these transient helices are known to interact with partners, whereas others represent putative binding sites for yet unidentified proteins. Finally, using NMR spectroscopy and isothermal titration calorimetry, we map a region of the phosphoprotein, comprising residues between 110 and 140 and common to the V and W proteins, that binds with weak affinity to STAT1 and confirm the involvement of key amino acids of the viral protein in this interaction. This provides new, to our knowledge, insights into how the phosphoprotein and the nonstructural V and W proteins of NiV perform their multiple functions.


Assuntos
Vírus Nipah , Fosfoproteínas , Conformação Proteica , Proteínas Virais , Replicação Viral
7.
Emerg Infect Dis ; 26(1): 104-113, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31855143

RESUMO

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.


Assuntos
Quirópteros/virologia , Infecções por Henipavirus/veterinária , Vírus Nipah/patogenicidade , Animais , Camboja , Genoma Viral/genética , Infecções por Henipavirus/epidemiologia , Infecções por Henipavirus/virologia , Humanos , Vírus Nipah/genética , Filogenia , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Sequenciamento Completo do Genoma
8.
Viruses ; 11(1)2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30587835

RESUMO

Since the largest 2014⁻2016 Ebola virus disease outbreak in West Africa, understanding of Ebola virus infection has improved, notably the involvement of innate immune mediators. Amongst them, collectins are important players in the antiviral innate immune defense. A screening of Ebola glycoprotein (GP)-collectins interactions revealed the specific interaction of human surfactant protein D (hSP-D), a lectin expressed in lung and liver, two compartments where Ebola was found in vivo. Further analyses have demonstrated an involvement of hSP-D in the enhancement of virus infection in several in vitro models. Similar effects were observed for porcine SP-D (pSP-D). In addition, both hSP-D and pSP-D interacted with Reston virus (RESTV) GP and enhanced pseudoviral infection in pulmonary cells. Thus, our study reveals a novel partner of Ebola GP that may participate to enhance viral spread.


Assuntos
Ebolavirus/química , Glicoproteínas/química , Doença pelo Vírus Ebola/imunologia , Proteína D Associada a Surfactante Pulmonar/química , Animais , Chlorocebus aethiops , Colectinas/química , Ebolavirus/efeitos dos fármacos , Células HEK293 , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Ligação Proteica , Proteína D Associada a Surfactante Pulmonar/genética , Suínos , Células Vero , Proteínas Virais/química
9.
J Infect Dis ; 218(suppl_5): S666-S671, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30239745

RESUMO

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.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Glicolipídeos/farmacologia , Manosídeos/uso terapêutico , Animais , Chlorocebus aethiops , Ebolavirus/fisiologia , Humanos , Células Vero , Internalização do Vírus/efeitos dos fármacos
10.
Cell ; 174(4): 938-952.e13, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30096313

RESUMO

Antibodies are promising post-exposure therapies against emerging viruses, but which antibody features and in vitro assays best forecast protection are unclear. Our international consortium systematically evaluated antibodies against Ebola virus (EBOV) using multidisciplinary assays. For each antibody, we evaluated epitopes recognized on the viral surface glycoprotein (GP) and secreted glycoprotein (sGP), readouts of multiple neutralization assays, fraction of virions left un-neutralized, glycan structures, phagocytic and natural killer cell functions elicited, and in vivo protection in a mouse challenge model. Neutralization and induction of multiple immune effector functions (IEFs) correlated most strongly with protection. Neutralization predominantly occurred via epitopes maintained on endosomally cleaved GP, whereas maximal IEF mapped to epitopes farthest from the viral membrane. Unexpectedly, sGP cross-reactivity did not significantly influence in vivo protection. This comprehensive dataset provides a rubric to evaluate novel antibodies and vaccine responses and a roadmap for therapeutic development for EBOV and related viruses.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Ebolavirus/imunologia , Epitopos/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Glicoproteínas de Membrana/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Feminino , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Resultado do Tratamento
11.
Antiviral Res ; 150: 183-192, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29305306

RESUMO

There are no approved medications for the treatment of Marburg or Ebola virus infection. In two previous articles (Martin et al., 2016, Martin et al., 2017), we reviewed surface glycoprotein and replication proteins structure/function relationship to decipher the molecular mechanisms of filovirus life cycle and identify antiviral strategies. In the present article, we recapitulate knowledge about the viral proteins involved in filovirus assembly and budding. First we describe the structural data available for viral proteins associated with virus assembly and virion egress and then, we integrate the structural features of these proteins in the functional context of the viral replication cycle. Finally, we summarize recent advances in the development of innovative antiviral strategies to target filovirus assembly and egress. The development of such prophylactic or post-exposure treatments could help controlling future filovirus outbreaks.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas , Filoviridae/efeitos dos fármacos , Filoviridae/fisiologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Montagem de Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Antivirais/química , Descoberta de Drogas/métodos , Filoviridae/classificação , Genoma Viral , Genômica/métodos , Humanos , Relação Estrutura-Atividade , Proteínas Virais/antagonistas & inibidores
12.
PLoS Pathog ; 13(9): e1006610, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28957419

RESUMO

IFITMs are broad antiviral factors that block incoming virions in endosomal vesicles, protecting target cells from infection. In the case of HIV-1, we and others reported the existence of an additional antiviral mechanism through which IFITMs lead to the production of virions of reduced infectivity. However, whether this second mechanism of inhibition is unique to HIV or extends to other viruses is currently unknown. To address this question, we have analyzed the susceptibility of a broad spectrum of viruses to the negative imprinting of the virion particles infectivity by IFITMs. The results we have gathered indicate that this second antiviral property of IFITMs extends well beyond HIV and we were able to identify viruses susceptible to the three IFITMs altogether (HIV-1, SIV, MLV, MPMV, VSV, MeV, EBOV, WNV), as well as viruses that displayed a member-specific susceptibility (EBV, DUGV), or were resistant to all IFITMs (HCV, RVFV, MOPV, AAV). The swapping of genetic elements between resistant and susceptible viruses allowed us to point to specificities in the viral mode of assembly, rather than glycoproteins as dominant factors of susceptibility. However, we also show that, contrarily to X4-, R5-tropic HIV-1 envelopes confer resistance against IFITM3, suggesting that viral receptors add an additional layer of complexity in the IFITMs-HIV interplay. Lastly, we show that the overall antiviral effects ascribed to IFITMs during spreading infections, are the result of a bimodal inhibition in which IFITMs act both by protecting target cells from incoming viruses and in driving the production of virions of reduced infectivity. Overall, our study reports for the first time that the negative imprinting of the virion particles infectivity is a conserved antiviral property of IFITMs and establishes IFITMs as a paradigm of restriction factor capable of interfering with two distinct phases of a virus life cycle.


Assuntos
Antígenos de Diferenciação/metabolismo , Vírion , Replicação Viral , Linhagem Celular , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Internalização do Vírus
13.
Viruses ; 9(5)2017 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-28492506

RESUMO

The mononegaviral family Filoviridae has eight members assigned to three genera and seven species. Until now, genus and species demarcation were based on arbitrarily chosen filovirus genome sequence divergence values (≈50% for genera, ≈30% for species) and arbitrarily chosen phenotypic virus or virion characteristics. Here we report filovirus genome sequence-based taxon demarcation criteria using the publicly accessible PAirwise Sequencing Comparison (PASC) tool of the US National Center for Biotechnology Information (Bethesda, MD, USA). Comparison of all available filovirus genomes in GenBank using PASC revealed optimal genus demarcation at the 55-58% sequence diversity threshold range for genera and at the 23-36% sequence diversity threshold range for species. Because these thresholds do not change the current official filovirus classification, these values are now implemented as filovirus taxon demarcation criteria that may solely be used for filovirus classification in case additional data are absent. A near-complete, coding-complete, or complete filovirus genome sequence will now be required to allow official classification of any novel "filovirus." Classification of filoviruses into existing taxa or determining the need for novel taxa is now straightforward and could even become automated using a presented algorithm/flowchart rooted in RefSeq (type) sequences.


Assuntos
Filoviridae/classificação , Filoviridae/genética , Filogenia , Algoritmos , Sequência de Bases , Bases de Dados de Ácidos Nucleicos , Ebolavirus/classificação , Ebolavirus/genética , Variação Genética , Genoma Viral , Marburgvirus/classificação , Marburgvirus/genética , Mononegavirais/classificação , Mononegavirais/genética , Análise de Sequência de DNA , Design de Software , Especificidade da Espécie , Sequenciamento Completo do Genoma
14.
Curr Opin Virol ; 22: 51-58, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28012412

RESUMO

Ever since the first recognised outbreak of Ebolavirus in 1976, retrospective epidemiological analyses and extensive studies with animal models have given us insight into the nature of the pathology and transmission mechanisms of this virus. In this review focusing on Ebolavirus, we present an outline of our current understanding of filovirus human-to-human transmission and of our knowledge concerning the molecular basis of viral transmission and potential for adaptation, with particular focus on what we have learnt from the 2014 outbreak in West Africa. We identify knowledge gaps relating to transmission and pathogenicity mechanisms, molecular adaptation and filovirus ecology.


Assuntos
Transmissão de Doença Infecciosa , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/transmissão , Humanos
15.
PLoS One ; 11(6): e0156775, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27280712

RESUMO

Polyclonal xenogenic IgGs, although having been used in the prevention and cure of severe infectious diseases, are highly immunogenic, which may restrict their usage in new applications such as Ebola hemorrhagic fever. IgG glycans display powerful xenogeneic antigens in humans, for example α1-3 Galactose and the glycolyl form of neuraminic acid Neu5Gc, and IgGs deprived of these key sugar epitopes may represent an advantage for passive immunotherapy. In this paper, we explored whether low immunogenicity IgGs had a protective effect on a guinea pig model of Ebola virus (EBOV) infection. For this purpose, a double knock-out pig lacking α1-3 Galactose and Neu5Gc was immunized against virus-like particles displaying surface EBOV glycoprotein GP. Following purification from serum, hyper-immune polyclonal IgGs were obtained, exhibiting an anti-EBOV GP titer of 1:100,000 and a virus neutralizing titer of 1:100. Guinea pigs were injected intramuscularly with purified IgGs on day 0 and day 3 post-EBOV infection. Compared to control animals treated with IgGs from non-immunized double KO pigs, the anti-EBOV IgGs-treated animals exhibited a significantly prolonged survival and a decreased virus load in blood on day 3. The data obtained indicated that IgGs lacking α1-3 Galactose and Neu5Gc, two highly immunogenic epitopes in humans, have a protective effect upon EBOV infection.


Assuntos
Anticorpos Antivirais/sangue , Vacinas contra Ebola/uso terapêutico , Galactose/deficiência , Doença pelo Vírus Ebola/prevenção & controle , Imunoglobulina G/imunologia , Ácidos Neuramínicos/metabolismo , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Anti-Idiotípicos/imunologia , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Cobaias , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/imunologia , Masculino , Suínos , Vacinação , Carga Viral
16.
J Virol ; 90(11): 5256-5269, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26984723

RESUMO

UNLABELLED: Ebola virus infection requires the surface viral glycoprotein to initiate entry into the target cells. The trimeric glycoprotein is a highly glycosylated viral protein which has been shown to interact with host C-type lectin receptors and the soluble complement recognition protein mannose-binding lectin, thereby enhancing viral infection. Similarly to mannose-binding lectin, ficolins are soluble effectors of the innate immune system that recognize particular glycans at the pathogen surface. In this study, we demonstrate that ficolin-1 interacts with the Zaire Ebola virus (EBOV) glycoprotein, and we characterized this interaction by surface plasmon resonance spectroscopy. Ficolin-1 was shown to bind to the viral glycoprotein with a high affinity. This interaction was mediated by the fibrinogen-like recognition domain of ficolin-1 and the mucin-like domain of the viral glycoprotein. Using a ficolin-1 control mutant devoid of sialic acid-binding capacity, we identified sialylated moieties of the mucin domain to be potential ligands on the glycoprotein. In cell culture, using both pseudotyped viruses and EBOV, ficolin-1 was shown to enhance EBOV infection independently of the serum complement. We also observed that ficolin-1 enhanced EBOV infection on human monocyte-derived macrophages, described to be major viral target cells,. Competition experiments suggested that although ficolin-1 and mannose-binding lectin recognized different carbohydrate moieties on the EBOV glycoprotein, the observed enhancement of the infection likely depended on a common cellular receptor/partner. In conclusion, ficolin-1 could provide an alternative receptor-mediated mechanism for enhancing EBOV infection, thereby contributing to viral subversion of the host innate immune system. IMPORTANCE: A specific interaction involving ficolin-1 (M-ficolin), a soluble effector of the innate immune response, and the glycoprotein (GP) of EBOV was identified. Ficolin-1 enhanced virus infection instead of tipping the balance toward its elimination. An interaction between the fibrinogen-like recognition domain of ficolin-1 and the mucin-like domain of Ebola virus GP occurred. In this model, the enhancement of infection was shown to be independent of the serum complement. The facilitation of EBOV entry into target host cells by the interaction with ficolin-1 and other host lectins shunts virus elimination, which likely facilitates the survival of the virus in infected host cells and contributes to the virus strategy to subvert the innate immune response.


Assuntos
Ebolavirus/metabolismo , Lectinas/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Mucinas/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Proteínas do Sistema Complemento/metabolismo , Ebolavirus/química , Ebolavirus/genética , Células HEK293 , Humanos , Macrófagos/virologia , Lectina de Ligação a Manose/metabolismo , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Células Vero , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Ficolinas
17.
Viruses ; 7(12): 6233-40, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26633464

RESUMO

The current outbreak of Ebola virus (EBOV) in West Africa has claimed the lives of more than 15,000 people and highlights an urgent need for therapeutics capable of preventing virus replication. In this study we screened known nucleoside analogues for their ability to interfere with EBOV replication. Among them, the cytidine analogue ß-d-N4-hydroxycytidine (NHC) demonstrated potent inhibitory activities against EBOV replication and spread at non-cytotoxic concentrations. Thus, NHC constitutes an interesting candidate for the development of a suitable drug treatment against EBOV.


Assuntos
Antivirais/farmacologia , Citidina/análogos & derivados , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/toxicidade , Chlorocebus aethiops , Citidina/farmacologia , Citidina/toxicidade , Células Vero
18.
J Infect Dis ; 212 Suppl 2: S372-8, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26232760

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Ebolavirus/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Glicoproteínas/imunologia , Células HEK293 , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Humanos , Ligação Proteica/imunologia , Células Vero , Vírion/imunologia , Internalização do Vírus
19.
J Infect Dis ; 212 Suppl 2: S226-33, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26138826

RESUMO

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.


Assuntos
Ebolavirus/genética , Genes Virais/genética , Doença pelo Vírus Ebola/virologia , Edição de RNA/genética , Proteínas do Envelope Viral/genética , Proteínas Virais/genética , Fatores de Virulência/genética , Animais , Linhagem Celular , Regulação para Baixo/genética , Ebolavirus/patogenicidade , Regulação Viral da Expressão Gênica/genética , Cobaias , Glicoproteínas de Membrana/genética , Mutação/genética
20.
J Infect Dis ; 212 Suppl 2: S322-8, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26092855

RESUMO

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.


Assuntos
Ebolavirus/metabolismo , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/virologia , Glicoproteínas de Membrana/metabolismo , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Ebolavirus/genética , Glicoproteínas de Membrana/genética , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/genética , Vírion/metabolismo , Vírion/patogenicidade , Virulência/genética , Internalização do Vírus , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...