Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Virology ; 567: 65-76, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35032865

RESUMO

Rift Valley fever virus (RVFV) is an arbovirus that was first reported in the Rift Valley of Kenya which causes significant disease in humans and livestock. RVFV is a tri-segmented, negative-sense RNA virus consisting of a L, M, and S segments with the M segment encoding the glycoproteins Gn and Gc. Host factors that interact with Gn are largely unknown. To this end, two viruses containing an epitope tag (V5) on the Gn protein in position 105 or 229 (V5Gn105 and V5Gn229) were generated using the RVFV MP-12 vaccine strain as a backbone. The V5-tag insertion minimally impacted Gn functionality as measured by replication kinetics, Gn localization, and antibody neutralization assays. A proteomics-based approach was used to identify novel Gn-binding host proteins, including the E3 ubiquitin-protein ligase, UBR4. Depletion of UBR4 resulted in a significant decrease in RVFV titers and a reduction in viral RNA production.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Interações Hospedeiro-Patógeno/genética , Vírus da Febre do Vale do Rift/genética , Ubiquitina-Proteína Ligases/genética , Proteínas do Envelope Viral/genética , Animais , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Culex , Epitopos/química , Epitopos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células HEK293 , Hepatócitos/virologia , Humanos , Ligação Proteica , Vírus da Febre do Vale do Rift/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Proteínas do Envelope Viral/metabolismo , Replicação Viral
2.
Nat Commun ; 11(1): 3281, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32612175

RESUMO

Amyloid fibrils result from the aggregation of host cell-encoded proteins, many giving rise to specific human illnesses such as Alzheimer's disease. Here we show that the major virulence factor of Rift Valley fever virus, the protein NSs, forms filamentous structures in the brain of mice and affects mortality. NSs assembles into nuclear and cytosolic disulfide bond-dependent fibrillary aggregates in infected cells. NSs structural arrangements exhibit characteristics typical for amyloids, such as an ultrastructure of 12 nm-width fibrils, a strong detergent resistance, and interactions with the amyloid-binding dye Thioflavin-S. The assembly dynamics of viral amyloid-like fibrils can be visualized in real-time. They form spontaneously and grow in an amyloid fashion within 5 hours. Together, our results demonstrate that viruses can encode amyloid-like fibril-forming proteins and have strong implications for future research on amyloid aggregation and toxicity in general.


Assuntos
Amiloide/metabolismo , Proteínas Amiloidogênicas/metabolismo , Febre do Vale de Rift/metabolismo , Vírus da Febre do Vale do Rift/metabolismo , Proteínas não Estruturais Virais/metabolismo , Amiloide/química , Amiloide/ultraestrutura , Proteínas Amiloidogênicas/química , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Chlorocebus aethiops , Células HeLa , Humanos , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Agregação Patológica de Proteínas/metabolismo , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/patogenicidade , Células Vero , Proteínas não Estruturais Virais/química , Virulência , Fatores de Virulência
3.
Biotechnol Lett ; 42(4): 529-536, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31983039

RESUMO

OBJECTIVES: The aim of the current study was to develop biodegradable alginate (ALG)/poly-L-lysine (PLL) microcapsules (MC) with entrapped plasmids expressing Gn and Gc glycoproteins of Rift Valley Fever virus (RVFV) and to evaluate the humoral immune response in mice. RESULTS: Expressing phRVF/Gn and phRVF/Gc plasmids which encode full-sized Gn and Gc glycoproteins and contain signal fusion protein F sequences of human parainfluenza (HPIV-1) were constructed. To protect the plasmids from cleavage by extracellular nucleases, they were entrapped into multilayer ALG/PLL microcapsules by layer-by-layer technique. To study the efficacy of humoral immune response, both native and microencapsulated plasmids were injected intramuscular into BALB/c mice. The humoral response in the mice immunized with free plasmids was characterized by virus-neutralizing antibody induction (with titres 1:4 to 1:8), while the injection of microencapsulated plasmids allowed to increase the titre level (from 1:16 to 1:32). CONCLUSION: The plasmids microencapsulated in biodegradable MC could be promising for development of DNA vaccines against RVFV.


Assuntos
Anticorpos Neutralizantes/metabolismo , Vetores Genéticos/administração & dosagem , Glicoproteínas/imunologia , Vírus da Febre do Vale do Rift/metabolismo , Alginatos/química , Animais , Anticorpos Antivirais/metabolismo , Cápsulas , Feminino , Vetores Genéticos/imunologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Imunidade Humoral , Imunização , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , Polilisina/análogos & derivados , Polilisina/química , Vírus da Febre do Vale do Rift/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
4.
J Virol ; 94(7)2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-31941775

RESUMO

Mosquito-borne La Crosse virus (LACV; genus Orthobunyavirus, family Peribunyaviridae, order Bunyavirales) causes up to 100 annual cases of severe meningoencephalitis in children and young adults in the United States. A major virulence factor of LACV is the nonstructural protein NSs, which inhibits host cell mRNA synthesis to prevent the induction of antiviral type I interferons (IFN-α/ß). To achieve this host transcriptional shutoff, LACV NSs drives the proteasomal degradation of RPB1, the large subunit of mammalian RNA polymerase II. Here, we show that NSs acts in a surprisingly rapid manner, as RPB1 degradation was commencing already at 1 h postinfection. The RPB1 degradation was partially dependent on the cellular E3 ubiquitin ligase subunit Elongin C. Consequently, removal of Elongin C, but also of the subunits Elongin A or B by siRNA transfection partially rescued general RNAP II transcription and IFN-beta mRNA synthesis from the blockade by NSs. In line with these results, LACV NSs was found to trigger the redistribution of Elongin C out of nucleolar speckles, which, however, is an epiphenomenon rather than part of the NSs mechanism. Our study also shows that the molecular phenotype of LACV NSs is different from RNA polymerase II inhibitors like α-amanitin or Rift Valley fever virus NSs, indicating that LACV is unique in involving the Elongin complex to shut off host transcription and IFN response.IMPORTANCE The mosquito-borne La Crosse virus (LACV; genus Orthobunyavirus, family Peribunyaviridae, order Bunyavirales) is prevalent in the United States and can cause severe childhood meningoencephalitis. Its main virulence factor, the nonstructural protein NSs, is a strong inhibitor of the antiviral type I interferon (IFN) system. NSs acts by imposing a global host mRNA synthesis shutoff, mediated by NSs-driven proteasomal degradation of the RPB1 subunit of RNA polymerase II. Here, we show that RPB1 degradation commences as early as 1 h postinfection, and identify the E3 ubiquitin ligase subunit Elongin C (and its binding partners Elongins A and B) as an NSs cofactor involved in RPB1 degradation and in suppression of global as well as IFN-related mRNA synthesis.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Elonguina/metabolismo , Vírus La Crosse/enzimologia , Proteínas não Estruturais Virais/metabolismo , Células A549 , Alfa-Amanitina/metabolismo , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Interferons/metabolismo , Vírus La Crosse/genética , Fenótipo , RNA Interferente Pequeno/metabolismo , Vírus da Febre do Vale do Rift/metabolismo , Transcrição Gênica , Células Vero , Fatores de Virulência/metabolismo
5.
BMC Biotechnol ; 18(1): 77, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30537953

RESUMO

BACKGROUND: Rift Valley fever virus (RVFV), the causative agent of Rift Valley fever, is an enveloped single-stranded negative-sense RNA virus in the genus Phlebovirus, family Bunyaviridae. The virus is spread by infected mosquitoes and affects ruminants and humans, causing abortion storms in pregnant ruminants, high neonatal mortality in animals, and morbidity and occasional fatalities in humans. The disease is endemic in parts of Africa and the Arabian Peninsula, but is described as emerging due to the wide range of mosquitoes that could spread the disease into non-endemic regions. There are different tests for determining whether animals are infected with or have been exposed to RVFV. The most common serological test is antibody ELISA, which detects host immunoglobulins M or G produced specifically in response to infection with RVFV. The presence of antibodies to RVFV nucleocapsid protein (N-protein) is among the best indicators of RVFV exposure in animals. This work describes an investigation of the feasibility of producing a recombinant N-protein in Nicotiana benthamiana and using it in an ELISA. RESULTS: The human-codon optimised RVFV N-protein was successfully expressed in N. benthamiana via Agrobacterium-mediated infiltration of leaves. The recombinant protein was detected as monomers and dimers with maximum protein yields calculated to be 500-558 mg/kg of fresh plant leaves. The identity of the protein was confirmed by liquid chromatography-mass spectrometry (LC-MS) resulting in 87.35% coverage, with 264 unique peptides. Transmission electron microscopy revealed that the protein forms ring structures of ~ 10 nm in diameter. Preliminary data revealed that the protein could successfully differentiate between sera of RVFV-infected sheep and from sera of those not infected with the virus. CONCLUSIONS: To the best of our knowledge this is the first study demonstrating the successful production of RVFV N-protein as a diagnostic reagent by Agrobacterium-mediated transient heterologous expression in N. benthamiana. Preliminary testing of the antigen showed its ability to distinguish RVFV-positive animal sera from RVFV negative animal sera when used in an enzyme linked immunosorbent assay (ELISA). The cost-effective, scalable and simple production method has great potential for use in developing countries where rapid diagnosis of RVFV is necessary.


Assuntos
Antígenos Virais/genética , Nicotiana/genética , Proteínas do Nucleocapsídeo/genética , Febre do Vale de Rift/diagnóstico , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/metabolismo , Doenças dos Ovinos/diagnóstico , Animais , Antígenos Virais/sangue , Antígenos Virais/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Expressão Gênica , Proteínas do Nucleocapsídeo/sangue , Proteínas do Nucleocapsídeo/metabolismo , Febre do Vale de Rift/sangue , Febre do Vale de Rift/virologia , Ovinos , Doenças dos Ovinos/sangue , Doenças dos Ovinos/virologia , Nicotiana/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(36): E7564-E7573, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28827346

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) and Rift Valley fever virus (RVFV) are two arthropod-borne phleboviruses in the Bunyaviridae family, which cause severe illness in humans and animals. Glycoprotein N (Gn) is one of the envelope proteins on the virus surface and is a major antigenic component. Despite its importance for virus entry and fusion, the molecular features of the phleboviruse Gn were unknown. Here, we present the crystal structures of the Gn head domain from both SFTSV and RVFV, which display a similar compact triangular shape overall, while the three subdomains (domains I, II, and III) making up the Gn head display different arrangements. Ten cysteines in the Gn stem region are conserved among phleboviruses, four of which are responsible for Gn dimerization, as revealed in this study, and they are highly conserved for all members in Bunyaviridae Therefore, we propose an anchoring mode on the viral surface. The complex structure of the SFTSV Gn head and human neutralizing antibody MAb 4-5 reveals that helices α6 in subdomain III is the key component for neutralization. Importantly, the structure indicates that domain III is an ideal region recognized by specific neutralizing antibodies, while domain II is probably recognized by broadly neutralizing antibodies. Collectively, Gn is a desirable vaccine target, and our data provide a molecular basis for the rational design of vaccines against the diseases caused by phleboviruses and a model for bunyavirus Gn embedding on the viral surface.


Assuntos
Anticorpos Neutralizantes/metabolismo , Epitopos/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Phlebovirus/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Infecções por Bunyaviridae/virologia , Linhagem Celular , Cristalografia por Raios X , Epitopos/química , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/metabolismo , Células Sf9 , Internalização do Vírus
7.
Sci Rep ; 6: 20617, 2016 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-26847478

RESUMO

Rift Valley Fever virus (RVFV) causes recurrent outbreaks of acute life-threatening human and livestock illness in Africa and the Arabian Peninsula. No licensed vaccines are currently available for humans and those widely used in livestock have major safety concerns. A 'One Health' vaccine development approach, in which the same vaccine is co-developed for multiple susceptible species, is an attractive strategy for RVFV. Here, we utilized a replication-deficient chimpanzee adenovirus vaccine platform with an established human and livestock safety profile, ChAdOx1, to develop a vaccine for use against RVFV in both livestock and humans. We show that single-dose immunization with ChAdOx1-GnGc vaccine, encoding RVFV envelope glycoproteins, elicits high-titre RVFV-neutralizing antibody and provides solid protection against RVFV challenge in the most susceptible natural target species of the virus-sheep, goats and cattle. In addition we demonstrate induction of RVFV-neutralizing antibody by ChAdOx1-GnGc vaccination in dromedary camels, further illustrating the potency of replication-deficient chimpanzee adenovirus vaccine platforms. Thus, ChAdOx1-GnGc warrants evaluation in human clinical trials and could potentially address the unmet human and livestock vaccine needs.


Assuntos
Vacinas contra Adenovirus/administração & dosagem , Anticorpos Neutralizantes/metabolismo , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/metabolismo , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas contra Adenovirus/farmacologia , Animais , Camelus , Bovinos , Cabras , Humanos , Pan troglodytes/imunologia , Pan troglodytes/virologia , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/imunologia , Arábia Saudita/epidemiologia , Ovinos , Reino Unido/epidemiologia , Vacinação , Vacinas Sintéticas/administração & dosagem , Proteínas do Envelope Viral/imunologia
8.
J Vis Exp ; (89): e51415, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-25146252

RESUMO

Host defenses to virus infection are dependent on a rapid detection by pattern recognition receptors (PRRs) of the innate immune system. In the cytoplasm, the PRRs RIG-I and PKR bind to specific viral RNA ligands. This first mediates conformational switching and oligomerization, and then enables activation of an antiviral interferon response. While methods to measure antiviral host gene expression are well established, methods to directly monitor the activation states of RIG-I and PKR are only partially and less well established. Here, we describe two methods to monitor RIG-I and PKR stimulation upon infection with an established interferon inducer, the Rift Valley fever virus mutant clone 13 (Cl 13). Limited trypsin digestion allows to analyze alterations in protease sensitivity, indicating conformational changes of the PRRs. Trypsin digestion of lysates from mock infected cells results in a rapid degradation of RIG-I and PKR, whereas Cl 13 infection leads to the emergence of a protease-resistant RIG-I fragment. Also PKR shows a virus-induced partial resistance to trypsin digestion, which coincides with its hallmark phosphorylation at Thr 446. The formation of RIG-I and PKR oligomers was validated by native polyacrylamide gel electrophoresis (PAGE). Upon infection, there is a strong accumulation of RIG-I and PKR oligomeric complexes, whereas these proteins remained as monomers in mock infected samples. Limited protease digestion and native PAGE, both coupled to western blot analysis, allow a sensitive and direct measurement of two diverse steps of RIG-I and PKR activation. These techniques are relatively easy and quick to perform and do not require expensive equipment.


Assuntos
RNA Helicases DEAD-box/metabolismo , RNA Viral/metabolismo , Vírus da Febre do Vale do Rift/metabolismo , eIF-2 Quinase/metabolismo , Western Blotting , Linhagem Celular , Proteína DEAD-box 58 , Eletroforese em Gel de Poliacrilamida , Humanos , Receptores Imunológicos , Vírus da Febre do Vale do Rift/genética
9.
PLoS One ; 9(5): e93483, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24809507

RESUMO

Rift Valley fever is a potentially fatal disease of humans and domestic animals caused by Rift Valley fever virus (RVFV). Infection with RVFV in ruminants can cause near 100% abortion rates and recent outbreaks in naïve human populations have suggested case fatality rates of greater than thirty percent. To elucidate the roles that host proteins play during RVFV infection, proteomic analysis of RVFV virions was conducted using complementary analytical approaches, followed by functional validation studies of select identified host factors. Coupling the more traditional Gel LC/MS/MS approach (SDS PAGE followed by liquid chromatography tandem mass spectrometry) with an alternative technique that preserves protein complexes allowed the protein complement of these viral particles to be thoroughly examined. In addition to viral proteins present within the virions and virion-associated host proteins, multiple macromolecular complexes were identified. Bioinformatic analysis showed that host chaperones were among over-represented protein families associated with virions, and functional experiments using siRNA gene silencing and small molecule inhibitors identified several of these heat shock proteins, including heat shock protein 90 (HSP90), as important viral host factors. Further analysis indicated that HSP inhibition effects occur during the replication/transcription phase of the virus life cycle, leading to significant lowering of viral titers without compromising the functional capacity of released virions. Overall, these studies provide much needed further insight into interactions between RVFV and host cells, increasing our understanding of the infection process and suggesting novel strategies for anti-viral development. In particular, considering that several HSP90 inhibitors have been advancing through clinical trials for cancer treatment, these results also highlight the exciting potential of repurposing HSP90 inhibitors to treat RVF.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico/metabolismo , Vírus da Febre do Vale do Rift/metabolismo , Proteínas Virais/metabolismo , Vírion/metabolismo , Inativação Gênica , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico/genética , Proteômica , RNA Interferente Pequeno , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/genética , Espectrometria de Massas em Tandem , Proteínas Virais/genética , Vírion/genética
10.
Virology ; 448: 1-14, 2014 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-24314631

RESUMO

The correct folding, heterodimerization and trafficking of Gn/Gc envelope glycoproteins of Rift Valley fever virus, RVFV (Bunyaviridae and Phlebovirus genus) are essential for Golgi assembly and budding of viral particles. The Gn and Gc carboxy-terminus contain a Golgi targeting and an ER-retrieval signal, respectively. We generated RVFV-like particles with mutations in the cytosolic tails of Gn or Gc and identified regions important for release of infectious particles. The role of specific amino-acids in these regions was further investigated by creating recombinant mutant viruses by reverse-genetics. Residues outside the suspected Golgi targeting motif, i.e. the di-lysine K29-K30 motif and the N43, R44 and I46 residues of the Gn cytosolic domain, appeared important for Golgi localization and RNP packaging. Concerning the Gc tail, replacement of K2 or K3 in the di-lysine motif, had a drastic impact on Gn trafficking and induced an important organelle redistribution and cell remodeling, greatly affecting particle formation and release.


Assuntos
Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/crescimento & desenvolvimento , Vírus da Febre do Vale do Rift/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular , Citosol/virologia , Complexo de Golgi/virologia , Humanos , Dados de Sequência Molecular , Vírus da Febre do Vale do Rift/genética , Proteínas do Envelope Viral/genética
11.
Proc Natl Acad Sci U S A ; 110(5): 1696-701, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23319635

RESUMO

Rift Valley fever virus (RVFV), like many other Bunyaviridae family members, is an emerging human and animal pathogen. Bunyaviruses have an outer lipid envelope bearing two glycoproteins, G(N) and G(C), required for cell entry. Bunyaviruses deliver their genome into the host-cell cytoplasm by fusing their envelope with an endosomal membrane. The molecular mechanism of this key entry step is unknown. The crystal structure of RVFV G(C) reveals a class II fusion protein architecture found previously in flaviviruses and alphaviruses. The structure identifies G(C) as the effector of membrane fusion and provides a direct view of the membrane anchor that initiates fusion. A structure of nonglycosylated G(C) reveals an extended conformation that may represent a fusion intermediate. Unanticipated similarities between G(C) and flavivirus envelope proteins reveal an evolutionary link between the two virus families and provide insights into the organization of G(C) in the outer shell of RVFV.


Assuntos
Glicoproteínas de Membrana/química , Estrutura Terciária de Proteína , Vírus da Febre do Vale do Rift/metabolismo , Proteínas do Envelope Viral/química , Aminoácidos/química , Animais , Cristalografia por Raios X , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Vírus da Febre do Vale do Rift/genética , Células Sf9 , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
12.
J Virol ; 86(24): 13642-52, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23035232

RESUMO

The entry of the enveloped Rift Valley fever virus (RVFV) into its host cell is mediated by the viral glycoproteins Gn and Gc. We investigated the RVFV entry process and, in particular, its pH-dependent activation mechanism using our recently developed nonspreading-RVFV-particle system. Entry of the virus into the host cell was efficiently inhibited by lysosomotropic agents that prevent endosomal acidification and by compounds that interfere with dynamin- and clathrin-dependent endocytosis. Exposure of plasma membrane-bound virions to an acidic pH (

Assuntos
Ácidos/metabolismo , Vírus da Febre do Vale do Rift/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Cricetinae , Primers do DNA , Drosophila , Eletroforese em Gel de Poliacrilamida , Endocitose , Citometria de Fluxo , Concentração de Íons de Hidrogênio , Microscopia de Fluorescência , Conformação Proteica , Proteínas Virais de Fusão/química
13.
J Biol Chem ; 287(40): 33198-214, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22847000

RESUMO

Rift Valley fever virus (RVFV) is an arbovirus that is classified as a select agent, an emerging infectious virus, and an agricultural pathogen. Understanding RVFV-host interactions is imperative to the design of novel therapeutics. Here, we report that an infection by the MP-12 strain of RVFV induces phosphorylation of the p65 component of the NFκB cascade. We demonstrate that phosphorylation of p65 (serine 536) involves phosphorylation of IκBα and occurs through the classical NFκB cascade. A unique, low molecular weight complex of the IKK-ß subunit can be observed in MP-12-infected cells, which we have labeled IKK-ß2. The IKK-ß2 complex retains kinase activity and phosphorylates an IκBα substrate. Inhibition of the IKK complex using inhibitors impairs viral replication, thus alluding to the requirement of an active IKK complex to the viral life cycle. Curcumin strongly down-regulates levels of extracellular infectious virus. Our data demonstrated that curcumin binds to and inhibits kinase activity of the IKK-ß2 complex in infected cells. Curcumin partially exerts its inhibitory influence on RVFV replication by interfering with IKK-ß2-mediated phosphorylation of the viral protein NSs and by altering the cell cycle of treated cells. Curcumin also demonstrated efficacy against ZH501, the fully virulent version of RVFV. Curcumin treatment down-regulated viral replication in the liver of infected animals. Our data point to the possibility that RVFV infection may result in the generation of novel versions of host components (such as IKK-ß2) that, by virtue of altered protein interaction and function, qualify as unique therapeutic targets.


Assuntos
Curcumina/farmacologia , NF-kappa B/antagonistas & inibidores , Vírus da Febre do Vale do Rift/metabolismo , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Regulação Viral da Expressão Gênica , Humanos , Quinase I-kappa B/metabolismo , Camundongos , Camundongos Transgênicos , Febre do Vale de Rift/virologia , Transcrição Gênica
14.
Virology ; 422(1): 6-12, 2012 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-22018491

RESUMO

Rift Valley fever virus (RVFV) causes significant morbidity and mortality in humans and livestock throughout Africa and the Middle East. The clinical disease ranges from mild febrile illness, to hepatitis, retinitis, encephalitis and fatal hemorrhagic fever. RVFV NSs protein has previously been shown to interfere in vitro with the interferon response, and RVFV lacking the NSs protein is attenuated in several animal models. Monocytes and macrophages are key players in the innate immune response via expression of various cytokines and chemokines. Here we demonstrate that wild-type RVFV infection of human monocyte-derived macrophages leads to a productive infection and inhibition of the innate immune response via decreased expression of IFN-α2, IFN-ß and TNF-α. Using a recombinant virus lacking the NSs protein, we show that this effect is mediated by the viral NSs protein. Finally, analysis of RVF patient samples demonstrated an association between a pro-inflammatory cytokine response and patient survival.


Assuntos
Citocinas/sangue , Macrófagos/imunologia , Macrófagos/virologia , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift , Proteínas não Estruturais Virais/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Citocinas/biossíntese , Feminino , Humanos , Imunidade Inata , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Masculino , Pessoa de Meia-Idade , RNA Viral/genética , RNA Viral/metabolismo , Febre do Vale de Rift/mortalidade , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/imunologia , Vírus da Febre do Vale do Rift/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Proteínas não Estruturais Virais/deficiência
15.
Virol J ; 5: 82, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18638365

RESUMO

BACKGROUND: Studies on Rift Valley Fever Virus (RVFV) infection process and morphogenesis have been hampered due to the biosafety conditions required to handle this virus, making alternative systems such as recombinant virus-like particles, that may facilitate understanding of these processes are highly desirable. In this report we present the expression and characterization of RVFV structural proteins N, Gn and Gc and demonstrate the efficient generation of RVFV virus-like particles (VLPs) using a baculovirus expression system. RESULTS: A recombinant baculovirus, expressing nucleocapsid (N) protein of RVFV at high level under the control of the polyhedrin promoter was generated. Gel filtration analysis indicated that expressed N protein could form complex multimers. Further, N protein complex when visualized by electron microscopy (EM) exhibited particulate, nucleocapsid like-particles (NLPs). Subsequently, a single recombinant virus was generated that expressed the RVFV glycoproteins (Gn/Gc) together with the N protein using a dual baculovirus vector. Both the Gn and Gc glycoproteins were detected not only in the cytoplasm but also on the cell surface of infected cells. Moreover, expression of the Gn/Gc in insect cells was able to induce cell-cell fusion after a low pH shift indicating the retention of their functional characteristics. In addition, assembly of these three structural proteins into VLPs was identified by purification of cells' supernatant through potassium tartrate-glycerol gradient centrifugation followed by EM analysis. The purified particles exhibited enveloped structures that were similar to the structures of the wild-type RVFV virion particle. In parallel, a second recombinant virus was constructed that expressed only Gc protein together with N protein. This dual recombinant virus also generated VLPs with clear spiky structures, but appeared to be more pleomorphic than the VLPs with both glycoproteins, suggesting that Gc and probably also Gn interacts with N protein complex independent of each other. CONCLUSION: Our results suggest that baculovirus expression system has enormous potential to produce large amount of VLPs that may be used both for fundamental and applied research of RVFV.


Assuntos
Proteínas Recombinantes/metabolismo , Vírus da Febre do Vale do Rift/genética , Proteínas Estruturais Virais/metabolismo , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Células Cultivadas , Glicoproteínas/genética , Glicoproteínas/metabolismo , Microscopia Eletrônica , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/metabolismo , Proteínas Recombinantes/genética , Vírus da Febre do Vale do Rift/metabolismo , Spodoptera , Proteínas Estruturais Virais/genética , Vírion/metabolismo , Virologia/métodos
16.
J Virol ; 81(24): 13335-45, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913816

RESUMO

Rift Valley fever virus (RVFV) is a member of the genus Phlebovirus within the family Bunyaviridae. It can cause severe epidemics among ruminants and fever, myalgia, a hemorrhagic syndrome, and/or encephalitis in humans. The RVFV M segment encodes the NSm and 78-kDa proteins and two major envelope proteins, Gn and Gc. The biological functions of the NSm and 78-kDa proteins are unknown; both proteins are dispensable for viral replication in cell cultures. To determine the biological functions of the NSm and 78-kDa proteins, we generated the mutant virus arMP-12-del21/384, carrying a large deletion in the pre-Gn region of the M segment. Neither NSm nor the 78-kDa protein was synthesized in arMP-12-del21/384-infected cells. Although arMP-12-del21/384 and its parental virus, arMP-12, showed similar growth kinetics and viral RNA and protein accumulation in infected cells, arMP-12-del21/384-infected cells induced extensive cell death and produced larger plaques than did arMP-12-infected cells. arMP-12-del21/384 replication triggered apoptosis, including the cleavage of caspase-3, the cleavage of its downstream substrate, poly(ADP-ribose) polymerase, and activation of the initiator caspases, caspase-8 and -9, earlier in infection than arMP-12. NSm expression in arMP-12-del21/384-infected cells suppressed the severity of caspase-3 activation. Further, NSm protein expression inhibited the staurosporine-induced activation of caspase-8 and -9, demonstrating that other viral proteins were dispensable for NSm's function in inhibiting apoptosis. RVFV NSm protein is the first identified Phlebovirus protein that has an antiapoptotic function.


Assuntos
Apoptose/efeitos dos fármacos , Vírus da Febre do Vale do Rift/patogenicidade , Proteínas não Estruturais Virais/farmacologia , Animais , Caspases/metabolismo , Linhagem Celular , Chlorocebus aethiops , Ativação Enzimática/efeitos dos fármacos , Humanos , Rim/citologia , Rim/virologia , Macrófagos/virologia , Camundongos , RNA Viral/análise , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/metabolismo , Vírus da Febre do Vale do Rift/fisiologia , Células Vero , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Ensaio de Placa Viral , Replicação Viral
17.
Virology ; 301(2): 226-35, 2002 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-12359425

RESUMO

Rift Valley Fever (RVF) virus is an arbovirus and is responsible for large outbreaks of disease predominantly in sub-Saharan Africa. However, several aspects of RVF virus transmission, such as high viremia, multiple vector species, and broad host range, result in a pathogen with high likelihood of geographic spread. RVF virus infection in humans and livestock is characterized by broad dissemination of RVF virus antigens throughout the body. We sought insight into the high pathogenicity and broad tropism of this virus through a characterization of its interaction with polarized epithelial cells. Our results indicate that infection and release of RVF virus in polarized epithelial cells occurs at both apical and basolateral membranes and hence is bidirectional. Furthermore, our results indicate that RVF virus causes disruptions in both the microfilament and the microtubule networks. These disruptions may provide a mechanism for bidirectional release of RVF virions.


Assuntos
Vírus da Febre do Vale do Rift/fisiologia , Citoesqueleto de Actina/fisiologia , Células CACO-2 , Membrana Celular/virologia , Polaridade Celular , Células Epiteliais/virologia , Humanos , Glicoproteínas de Membrana/metabolismo , Microtúbulos/fisiologia , Vírus da Febre do Vale do Rift/metabolismo , Proteínas do Envelope Viral/metabolismo
18.
J Gen Virol ; 77 ( Pt 12): 3053-62, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9000097

RESUMO

Infection of mammalian cells with Rift Valley fever virus (RVFV) leads generally to the production of virus and cell death. In this paper we examined the fate of Vero cells infected with three strains of RVFV and observed that, while a large proportion of cells exhibited a clear cytopathic effect (CPE), a small but significant fraction did not undergo a lytic infection but was able to proliferate and establish a persistent infection. Several independent RVFV persistently infected cell lines have been established and passaged for more than 1 year after infection with a virulent strain (ZH548) and two attenuated strains (C13 and MP12). Although the viruses used for the primary infection were plaque-purified, we do not know whether defective-interfering particles were responsible for the establishment of the persistent infection. The persistently infected cells became resistant to superinfection with RVFV but not with other viruses and shed low amounts of infectious, lytic and non-lytic virus during a limited number of passages. In all the passages tested, the three genomic segments or related products were synthesized as well as the structural nucleoprotein N and glycoproteins G1 and G2. Abnormal defective RNAs were detected, migrating faster or slower than their respective counterparts. The faster-migrating RNAs were internally deleted, some of them possessing only the very terminal part of the 5' genomic end.


Assuntos
Vírus da Febre do Vale do Rift/fisiologia , Latência Viral , Animais , Chlorocebus aethiops , Camundongos , RNA Viral/biossíntese , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/isolamento & purificação , Vírus da Febre do Vale do Rift/metabolismo , Células Vero , Proteínas Estruturais Virais/biossíntese
19.
J Biol Chem ; 266(25): 16420-4, 1991 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-1885574

RESUMO

One of the major physiologic functions of erythrocytes is the mediation of chloride-bicarbonate exchange in the transport of carbon dioxide from the tissues to the lungs. The anion exchange is mediated by a typical polytopic transmembrane protein in the cell membrane, designated Band 3. A carboxyl-terminal peptide of Band 3 was affinity-labeled with pyridoxal phosphate, a substrate for the anion transport system, and then sequenced (Kawano, Y., Okubo, K., Tokunaga, F., Miyata, T., Iwanaga, S., and Hamasaki, N. (1988) J. Biol. Chem. 263, 8232-8238). The 10th amino acid residue of the peptide could not be determined, suggesting post-translational modification of the residue. In the present communication, we have investigated the molecular structure of human Band 3 and the COOH-terminal 8500-dalton peptide using gas-liquid chromatography-mass spectrometry. Band 3 was modified covalently by fatty acids and these acids were released from Band 3 by hydroxylamine treatment at either pH 7 or 11, indicating that the linkage between Band 3 and the fatty acid is a thio ester bond. 1 mol of Band 3 interacted with 1 mol of fatty acid at a cysteine residue located 69 residues from the COOH terminus of Band 3. The fatty acids used in the modification were myristate, palmitate, oleate, and stearate, with palmitate being the major component. The esterified site is close to the site affinity-labeled with pyridoxal phosphate (Kawano, Y., Okubo, K., Tokunaga, F., Miyata, T., Iwanaga, S., and Hamasaki, N. (1988) J. Biol. Chem. 263, 8232-8238). The amino acid sequence including the acylation site was Phe-Thr-Gly-Ile-Gln-Ile-Ile-Cys-Leu-Ala-Val-Leu, which is conserved in the G2 protein of Rift Valley fever virus as Phe-Ser-Ser-Ile-Ala-Ile-Ile-Cys-Leu-Ala-Val-Leu. The G2 protein, like Band 3, is a polytopic transmembrane protein. Although acylation of the cysteine residue of G2 protein has not been examined, the Phe-X-X-Ile-X-Ile-Ile-Cys-Leu-Ala-Val-Leu sequence could be a common motif for fatty acylation of certain membrane proteins.


Assuntos
Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Cisteína/metabolismo , Membrana Eritrocítica/metabolismo , Ácidos Palmíticos/metabolismo , Vírus da Febre do Vale do Rift/metabolismo , Proteínas Virais/metabolismo , Acilação , Sequência de Aminoácidos , Sequência Consenso , Ácidos Graxos/metabolismo , Humanos , Hidroxilamina , Hidroxilaminas/farmacologia , Dados de Sequência Molecular , Alinhamento de Sequência
20.
Virology ; 170(2): 505-10, 1989 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2728348

RESUMO

The M segment RNA of Rift Valley fever virus (RVFV) encodes four gene products: the two viral envelop glycoproteins G2 and G1, a glycosylated 78-kDa protein, and a nonglycosylated 14-kDa protein. These proteins are generated from a single open reading frame (ORF) by a strategy involving independent translational initiations at both the first and second in-phase ATG codons and co-translational processing of primary polyprotein products. The ORF encodes six sites for N-linked glycosylation: one present in the "preglycoprotein region" preceding the coding sequences of the mature envelop glycoproteins, and within the coding sequences of both the 78- and 14-kDa proteins; one site in the glycoprotein G2 coding region, also present in the 78-kDa protein; and four sites within glycoprotein G1. From analyses of RVFV proteins produced in cells infected with recombinant vaccinia viruses expressing various M segment regions, we show glycoprotein G2 was glycosylated at its single site and glycoprotein G1 at at least three sites. Both sites for N-linked glycosylation in the 78-kDa protein were occupied with glycan. This latter result indicated the preglycoprotein region glycosylation site was utilized in the 78-kDa protein, but this same site within the 14-kDa protein was not. Further analysis showed utilization of this glycosylation site, as well as proteolytic processing at the amino terminus of the mature glycoprotein G2, appeared to be determined by initiation codon usage. The two-site translational initiation expression strategy of this phlebovirus M segment and its role in the control of post-translational protein modification and processing are discussed.


Assuntos
Bunyaviridae/genética , Regulação da Expressão Gênica , Glicoproteínas/genética , Vírus da Febre do Vale do Rift/genética , Proteínas Virais/genética , Glicoproteínas/metabolismo , Glicosilação , Testes de Precipitina , Vírus da Febre do Vale do Rift/metabolismo , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA