Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Appl Microbiol Biotechnol ; 108(1): 303, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38639795

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) causes the highly fatal disease in humans. To facilitate diagnosis, the native form of subunit glycoprotein (Gn), a prime target for potential vaccines and therapies, was produced in Nicotiana benthamiana using a Bamboo mosaic virus-based vector system. By fusion with secretory signal tags, SSExt, derived from the extension protein, and the (SP)10 motif, the yield of the recombinant Gn (rGn) was remarkably increased to approximately 7 mg/kg infiltrated leaves. Ultimately, an rGn-based ELISA was successfully established for the detection of SFTSV-specific antibodies in serum samples from naturally infected monkeys. As validated with the reference method, the specificity and sensitivity of rGn-ELISA were 94% and 96%, respectively. In conclusion, utilizing well-suited fusion tags facilitates rGn production and purification in substantial quantities while preserving its antigenic properties. The rGn-ELISA, characterized by its commendable sensitivity and specificity could serve as a viable alternative diagnostic method for assessing SFTSV seroprevalence. KEY POINTS: • SFTSV Gn, fused with secretory signal tags, was expressed by the BaMV-based vector. • The plant fusion tags increased expression levels and eased the purification of rGn. • The rGn-ELISA was established and validated; its specificity and sensitivity > 94%.


Assuntos
Phlebovirus , Febre Grave com Síndrome de Trombocitopenia , Humanos , Febre Grave com Síndrome de Trombocitopenia/diagnóstico , Phlebovirus/genética , Phlebovirus/metabolismo , Estudos Soroepidemiológicos , Glicoproteínas/metabolismo , Anticorpos
2.
Microb Pathog ; 178: 106079, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36966885

RESUMO

Experimental animal model is indispensable to evaluate the prophylactic and therapeutic candidates against severe fever with thrombocytopenia syndrome virus (SFTSV). To develop a suitable mouse model for SFTSV infection, we delivered human dendritic cell-specific ICAM-3-grabbing non-integrin (hDC-SIGN) by adeno-associated virus (AAV2) and validated its susceptibility for SFTSV infection. Western blot and RT-PCR assays confirmed the expression of hDC-SIGN in transduced cell lines and a significantly increased viral infectivity was observed in cells expressing hDC-SIGN. The C57BL/6 mice transduced with AAV2 exhibited a stable hDC-SIGN expression in the organs for 7 days. Upon SFTSV challenge with 1 × 105 FAID50, the mice transduced with rAAV-hDC-SIGN showed a 12.5% mortality and reduced platelet and white blood cell count in accordance with higher viral titer than control group. Liver and spleen samples collected from the transduced mice had pathological signs similar to the IFNAR-/- mice with severe SFTSV infection. Collectively, the rAAV-hDC-SIGN transduced mouse model can be used as an accessible and promising tool for studying the SFTSV pathogenesis and pre-clinical evaluation of vaccines and therapeutics against the SFTSV infection.


Assuntos
Phlebovirus , Febre Grave com Síndrome de Trombocitopenia , Humanos , Animais , Camundongos , Camundongos Endogâmicos C57BL , Phlebovirus/genética , Phlebovirus/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Modelos Animais de Doenças
3.
Sci Adv ; 8(19): eabj6894, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35544562

RESUMO

Endogenous viral elements (EVEs), accounting for 15% of our genome, serve as a genetic reservoir from which new genes can emerge. Nematode EVEs are particularly diverse and informative of virus evolution. We identify Atlas virus-an intact retrovirus-like EVE in the human hookworm Ancylostoma ceylanicum, with an envelope protein genetically related to GN-GC glycoproteins from the family Phenuiviridae. A cryo-EM structure of Atlas GC reveals a class II viral membrane fusion protein fold not previously seen in retroviruses. Atlas GC has the structural hallmarks of an active fusogen. Atlas GC trimers insert into membranes with endosomal lipid compositions and low pH. When expressed on the plasma membrane, Atlas GC has cell-cell fusion activity. With its preserved biological activities, Atlas GC has the potential to acquire a cellular function. Our work reveals structural plasticity in reverse-transcribing RNA viruses.


Assuntos
Phlebovirus , Vírus de RNA , Ancylostomatoidea/metabolismo , Animais , Humanos , Phlebovirus/química , Phlebovirus/genética , Phlebovirus/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus
4.
J Immunol ; 207(2): 590-601, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34244294

RESUMO

The nonstructural protein (NSs) of severe fever with thrombocytopenia syndrome virus (SFTSV) plays multiple functions in the virus life cycle. Proteomic screening for host proteins interacting with NSs identified the cellular protein LSm14A. LSm14A, a member of the LSm family involved in RNA processing in the processing bodies, binds to viral RNA or synthetic homolog and mediates IFN regulatory factor 3 activation and IFN-ß induction. NSs interacted with and colocalized with LSm14A, and this interaction effectively inhibited downstream phosphorylation and dimerization of IFN regulatory factor 3, resulting in the suppression of antiviral signaling and IFN induction in several cell types of human origin. Knockdown of NSs resulted in the suppression of SFTSV replication in host cells. Viral RNA bound to LSm14A-NSs protein complex during the interaction. A newly discovered LRRD motif of NSs functioned to interact with LSm14A. Altogether, our data demonstrated a mechanism used by SFTSV to inhibit host innate immune response.


Assuntos
Antivirais/metabolismo , Phlebovirus/metabolismo , Ribonucleoproteínas/metabolismo , Febre Grave com Síndrome de Trombocitopenia/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Imunidade Inata/fisiologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/fisiologia , Proteômica/métodos , Transdução de Sinais/fisiologia
5.
Nat Microbiol ; 5(6): 864-871, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32341479

RESUMO

Segmented negative-sense RNA viruses (sNSRVs) encode a single-polypeptide polymerase (L protein) or a heterotrimeric polymerase complex to cannibalize host messenger RNA cap structures serving as primers of transcription, and catalyse RNA synthesis. Here, we report the full-length structure of the severe fever with thrombocytopaenia syndrome virus (SFTSV) L protein, as determined by cryogenic electron microscopy at 3.4 Å, leading to an atomic model harbouring three functional parts (an endonuclease, an RNA-dependent RNA polymerase and a cap-binding domain) and two structural domains (an arm domain with a blocker motif and a carboxy-terminal lariat domain). The SFTSV L protein has a compact architecture in which its cap-binding pocket is surprisingly occupied by an Arg finger of the blocker motif, and the endonuclease active centre faces back towards the cap-binding pocket, suggesting that domain rearrangements are necessary to acquire the pre-initiation state of the active site. Our results provide insight into the complete architecture of sNSRV-encoded L protein and further the understanding of sNSRV transcription initiation.


Assuntos
Modelos Moleculares , Phlebovirus/genética , Phlebovirus/metabolismo , RNA Viral , Iniciação da Transcrição Genética , Proteínas Virais/química , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Sequência Conservada , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade , Transcrição Gênica
6.
PLoS One ; 14(10): e0223978, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31618247

RESUMO

Guertu virus (GTV) is a tick-borne phleboviruses (TBPVs) which belongs to the genus Banyangvirus in the family of Phenuiviridae. In vitro and in vivo studies of GTV demonstrated that it was able to infect animal and human cell lines and could cause pathological lesions in mice. Glycoproteins (GP, including Gn and Gc) on the surface of Guertu virus (GTV) could bind to receptors on host cells and induce protective immunity in the host, but knowledge is now lacking on the information of B cell epitopes (BCEs) present on GTV-GP protein. The aim of this study was to identify all BCEs on Gn of the GTV DXM strain using rabbit pAbs against GTV-Gn. Seven fine BCEs and two antigenic peptides (APs) from nine reactive 16mer-peptides were identified, which are EGn1 (2PIICEGLTHS11), EGn2 (135CSQDSGT141), EGn3 (165IP EDVF170), EGn4 (169VFQEL K174), EGn5 (187IDGILFN193), EGn6 (223QTKWIQ228), EGn7 (237CHKDGIGPC245), AP-8 (299GVRVRPKCYGFSRMMA314) and AP-9 (355CASH FCSSAESGKKNT370), of which six of mapped BCEs were recognized by the IgG-positive sheep serum obtained from sheep GTV-infected naturally. Multiple sequence alignments (MSA) based on each mapped BCE motif identified that the most of identified BCEs and APs are highly conserved among 10 SFTSV strains from different countries and lineages that share relatively close evolutionary relationships with GTV. The fine epitope mapping of the GTV-Gn would provide basic data with which to explore the GTV-Gn antigen structure and pathogenic mechanisms, and it could lay the foundation for the design and development of a GTV multi-epitope peptide vaccine and detection antigen.


Assuntos
Mapeamento de Epitopos/métodos , Glicoproteínas/química , Peptídeos/metabolismo , Phlebovirus/metabolismo , Sequência de Aminoácidos , Animais , Modelos Moleculares , Conformação Proteica , Coelhos , Alinhamento de Sequência , Ovinos/imunologia , Proteínas do Envelope Viral/química
7.
Biometals ; 31(1): 81-89, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29209895

RESUMO

Here we report on the results obtained from an antiviral screening, including herpes simplex virus, vaccinia virus, vesicular stomatitis virus, Coxsackie B4 virus or respiratory syncytial virus, parainfluenza-3 virus, reovirus-1 and Punta Toro virus, of three 2-hydroxy-3-methoxyphenyl acylhydrazone compounds in three cell lines (i.e. human embryonic lung fibroblast cells, human cervix carcinoma cells, and African Green monkey kidney cells). Interesting antiviral EC50 values are obtained against herpes simplex virus-1 and vaccinia virus. The biological activity of acylhydrazones is often attributed to their metal coordinating abilities, so potentiometric and microcalorimetric studies are here discussed to unravel the behavior of the three 2-hydroxy-3-methoxyphenyl compounds in solution. It is worth of note that the acylhydrazone with the higher affinity for Cu(II) ions shows the best antiviral activity against herpes simplex and vaccinia virus (EC50 ~ 1.5 µM, minimal cytotoxic concentration = 60 µM, selectivity index = 40).


Assuntos
Antivirais/farmacologia , Quelantes/farmacologia , Hidrazonas/farmacologia , Simplexvirus/efeitos dos fármacos , Vaccinia virus/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Quelantes/síntese química , Quelantes/metabolismo , Chlorocebus aethiops , Cobre/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Humanos , Hidrazonas/síntese química , Hidrazonas/metabolismo , Concentração Inibidora 50 , Magnésio/metabolismo , Manganês/metabolismo , Orthoreovirus de Mamíferos/efeitos dos fármacos , Orthoreovirus de Mamíferos/crescimento & desenvolvimento , Orthoreovirus de Mamíferos/metabolismo , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/metabolismo , Phlebovirus/efeitos dos fármacos , Phlebovirus/crescimento & desenvolvimento , Phlebovirus/metabolismo , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/metabolismo , Simplexvirus/crescimento & desenvolvimento , Simplexvirus/metabolismo , Vaccinia virus/crescimento & desenvolvimento , Vaccinia virus/metabolismo , Células Vero , Vesiculovirus/efeitos dos fármacos , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/metabolismo
8.
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
9.
Adv Virus Res ; 98: 83-118, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28433053

RESUMO

The Bunyavirales Order encompasses nine families of enveloped viruses containing a single-stranded negative-sense RNA genome divided into three segments. The small (S) and large (L) segments encode proteins participating in genome replication in the infected cell cytoplasm. The middle (M) segment encodes the viral glycoproteins Gn and Gc, which are derived from a precursor polyprotein by host cell proteases. Entry studies are available only for a few viruses in the Order, and in each case they were shown to enter cells via receptor-mediated endocytosis. The acidic endosomal pH triggers the fusion of the viral envelope with the membrane of an endosome. Structural studies on two members of this Order, the phleboviruses and the hantaviruses, have shown that the membrane fusion protein Gc displays a class II fusion protein fold and is homologous to its counterparts in flaviviruses and alphaviruses, which are positive-sense, single-stranded RNA viruses. We analyze here recent data on the structure and function of the structure of the phlebovirus Gc and hantavirus Gn and Gc glycoproteins, and extrapolate common features identified in the amino acid sequences to understand also the structure and function of their counterparts in other families of the Bunyavirales Order. Our analysis also identified clear structural homology between the hantavirus Gn and alphavirus E2 glycoproteins, which make a heterodimer with the corresponding fusion proteins Gc and E1, respectively, revealing that not only the fusion protein has been conserved across viral families.


Assuntos
Genoma Viral , Orthohantavírus/genética , Phlebovirus/genética , RNA Viral/genética , Proteínas Virais de Fusão/química , Alphavirus/classificação , Alphavirus/genética , Alphavirus/metabolismo , Alphavirus/ultraestrutura , Animais , Evolução Biológica , Infecções por Bunyaviridae/virologia , Endossomos/metabolismo , Endossomos/virologia , Orthohantavírus/classificação , Orthohantavírus/metabolismo , Orthohantavírus/ultraestrutura , Humanos , Modelos Moleculares , Phlebovirus/classificação , Phlebovirus/metabolismo , Phlebovirus/ultraestrutura , Filogenia , Dobramento de Proteína , RNA Viral/metabolismo , Homologia Estrutural de Proteína , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus
10.
Proc Natl Acad Sci U S A ; 113(26): 7154-9, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27325770

RESUMO

An emergent viral pathogen termed severe fever with thrombocytopenia syndrome virus (SFTSV) is responsible for thousands of clinical cases and associated fatalities in China, Japan, and South Korea. Akin to other phleboviruses, SFTSV relies on a viral glycoprotein, Gc, to catalyze the merger of endosomal host and viral membranes during cell entry. Here, we describe the postfusion structure of SFTSV Gc, revealing that the molecular transformations the phleboviral Gc undergoes upon host cell entry are conserved with otherwise unrelated alpha- and flaviviruses. By comparison of SFTSV Gc with that of the prefusion structure of the related Rift Valley fever virus, we show that these changes involve refolding of the protein into a trimeric state. Reverse genetics and rescue of site-directed histidine mutants enabled localization of histidines likely to be important for triggering this pH-dependent process. These data provide structural and functional evidence that the mechanism of phlebovirus-host cell fusion is conserved among genetically and patho-physiologically distinct viral pathogens.


Assuntos
Febre por Flebótomos/virologia , Phlebovirus/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Humanos , Phlebovirus/química , Phlebovirus/genética , Conformação Proteica , Alinhamento de Sequência , Proteínas do Envelope Viral/genética , Internalização do Vírus
11.
J Virol ; 89(6): 3026-37, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25552716

RESUMO

UNLABELLED: Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen that was first reported in China in 2009. Phylogenetic analysis of the viral genome showed that SFTS virus represents a new lineage within the Phlebovirus genus, distinct from the existing sandfly fever and Uukuniemi virus groups, in the family Bunyaviridae. SFTS disease is characterized by gastrointestinal symptoms, chills, joint pain, myalgia, thrombocytopenia, leukocytopenia, and some hemorrhagic manifestations with a case fatality rate of about 2 to 15%. Here we report the development of reverse genetics systems to study STFSV replication and pathogenesis. We developed and optimized functional T7 polymerase-based M- and S-segment minigenome assays, which revealed errors in the published terminal sequences of the S segment of the Hubei 29 strain of SFTSV. We then generated recombinant viruses from cloned cDNAs prepared to the antigenomic RNAs both of the minimally passaged virus (HB29) and of a cell culture-adapted strain designated HB29pp. The growth properties, pattern of viral protein synthesis, and subcellular localization of viral N and NSs proteins of wild-type HB29pp (wtHB29pp) and recombinant HB29pp viruses were indistinguishable. We also show that the viruses fail to shut off host cell polypeptide production. The robust reverse genetics system described will be a valuable tool for the design of therapeutics and the development of killed and attenuated vaccines against this important emerging pathogen. IMPORTANCE: SFTSV and related tick-borne phleboviruses such as Heartland virus are emerging viruses shown to cause severe disease in humans in the Far East and the United States, respectively. Study of these novel pathogens would be facilitated by technology to manipulate these viruses in a laboratory setting using reverse genetics. Here, we report the generation of infectious SFTSV from cDNA clones and demonstrate that the behavior of recombinant viruses is similar to that of the wild type. This advance will allow for further dissection of the roles of each of the viral proteins in the context of virus infection, as well as help in the development of antiviral drugs and protective vaccines.


Assuntos
Febre por Flebótomos/virologia , Phlebovirus/genética , Genética Reversa/métodos , Sequência de Aminoácidos , Sequência de Bases , China , Feminino , Genoma Viral , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Phlebovirus/química , Phlebovirus/metabolismo , Alinhamento de Sequência , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
12.
J Virol ; 88(8): 4572-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24478431

RESUMO

UNLABELLED: Recognition of viral pathogens by the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) family results in the activation of type I interferon (IFN) responses. To avoid this response, most viruses have evolved strategies that target different essential steps in the activation of host innate immunity. In this study, we report that the nonstructural protein NSs of the newly described severe fever with thrombocytopenia syndrome virus (SFTSV) is a potent inhibitor of IFN responses. The SFTSV NSs protein was found to inhibit the activation of the beta interferon (IFN-ß) promoter induced by viral infection and by a RIG-I ligand. Astonishingly, we found that SFTSV NSs interacts with and relocalizes RIG-I, the E3 ubiquitin ligase TRIM25, and TANK-binding kinase 1 (TBK1) into SFTSV NSs-induced cytoplasmic structures. Interestingly, formation of these SFTSV NSs-induced structures occurred in the absence of the Atg7 gene, a gene essential for autophagy. Furthermore, confocal microscopy studies revealed that these SFTSV NSs-induced structures colocalize with Rab5 but not with Golgi apparatus or endoplasmic reticulum markers. Altogether, the data suggest that sequestration of RIG-I signaling molecules into endosome-like structures may be the mechanism used by SFTSV to inhibit IFN responses and point toward a novel mechanism for the suppression of IFN responses. IMPORTANCE: The mechanism by which the newly described SFTSV inhibits host antiviral responses has not yet been fully characterized. In this study, we describe the redistribution of RIG-I signaling components into virus-induced cytoplasmic structures in cells infected with SFTSV. This redistribution correlates with the inhibition of host antiviral responses. Further characterization of the interplay between the viral protein and components of the IFN responses could potentially provide targets for the rational development of therapeutic interventions.


Assuntos
Infecções por Bunyaviridae/enzimologia , RNA Helicases DEAD-box/metabolismo , Endossomos/metabolismo , Interferon Tipo I/imunologia , Phlebovirus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/virologia , Linhagem Celular , Estruturas Citoplasmáticas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Endossomos/genética , Humanos , Interferon Tipo I/genética , Phlebovirus/genética , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Receptores Imunológicos , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas não Estruturais Virais/genética
13.
Microbiol Immunol ; 54(1): 20-30, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20055939

RESUMO

Punta Toro virus (PTV; family Bunyaviridae, genus Phlebovirus) causes severe hepatic damage through brisk apoptosis of hepatocytes. In the present study, two viral proteins encoded by the S segment of the viral genome, non-structural (NSs) and nucleocapsid protein (N), were examined for their roles in apoptosis. Expression of NSs in HepG2 cells led to apoptosis in 45% of transfected cells, and with N, 28%, on average. These levels represent a four- to an eightfold increase over cells transfected with the mutated protein vectors. Caspase-3, -8 and -9 activities were increased by N protein when compared with the control NC (P < 0.05), and by NSsA and NSsB, as compared to control NSsC (P < 0.01). Treatment of the transfected cells with caspase-8 or -9 inhibitors markedly decreased apoptosis. Neutralization of TNF-alpha or Fas ligand had no effect on apoptosis. These results indicate that both NSs and N are responsible for causing hepatocyte apoptosis by triggering the extrinsic caspase-8 and intrinsic caspase-9 pathways.


Assuntos
Apoptose/fisiologia , Hepatócitos/citologia , Proteínas do Nucleocapsídeo/fisiologia , Phlebovirus/metabolismo , Proteínas não Estruturais Virais/fisiologia , Animais , Apoptose/genética , Western Blotting , Caspase 8/metabolismo , Caspase 9/metabolismo , Chlorocebus aethiops , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Proteínas do Nucleocapsídeo/genética , Phlebovirus/genética , Reação em Cadeia da Polimerase , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células Vero , Proteínas não Estruturais Virais/genética
14.
Theor Biol Med Model ; 1: 10, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15544707

RESUMO

The Bunyaviridae family of enveloped RNA viruses includes five genuses, orthobunyaviruses, hantaviruses, phleboviruses, nairoviruses and tospoviruses. It has not been determined which Bunyavirus protein mediates virion:cell membrane fusion. Class II viral fusion proteins (beta-penetrenes), encoded by members of the Alphaviridae and Flaviviridae, are comprised of three antiparallel beta sheet domains with an internal fusion peptide located at the end of domain II. Proteomics computational analyses indicate that the carboxyl terminal glycoprotein (Gc) encoded by Sandfly fever virus (SAN), a phlebovirus, has a significant amino acid sequence similarity with envelope protein 1 (E1), the class II fusion protein of Sindbis virus (SIN), an Alphavirus. Similar sequences and common structural/functional motifs, including domains with a high propensity to interface with bilayer membranes, are located collinearly in SAN Gc and SIN E1. Gc encoded by members of each Bunyavirus genus share several sequence and structural motifs. These results suggest that Gc of Bunyaviridae, and similar proteins of Tenuiviruses and a group of Caenorhabditis elegans retroviruses, are class II viral fusion proteins. Comparisons of divergent viral fusion proteins can reveal features essential for virion:cell fusion, and suggest drug and vaccine strategies.


Assuntos
Alcenos/classificação , Bunyaviridae/metabolismo , Glicoproteínas/classificação , Modelos Biológicos , Proteômica , Proteínas do Envelope Viral/classificação , Proteínas Virais de Fusão/classificação , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/virologia , Sequência Conservada , Glicoproteínas/química , Glicoproteínas/genética , Dados de Sequência Molecular , Estrutura Molecular , Phlebovirus/metabolismo , Estrutura Terciária de Proteína , Retroviridae/metabolismo , Tenuivirus/metabolismo , Proteínas do Envelope Viral/química
15.
J Biol Chem ; 269(36): 22565-73, 1994 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-8077205

RESUMO

The G1 and G2 glycoproteins of Punta Toro virus, a member of the bunyaviruses, are targeted to the Golgi complex, where viral budding occurs. We found that the G1 protein, when expressed in the absence of G2, is also targeted to the Golgi complex. A series of G1 proteins truncated at the carboxyl-terminal region was constructed, and the localization of the expressed proteins was examined. It was found that the proteins expressed from constructs with partial deletions in the cytoplasmic domain were transported to the Golgi complex at a significantly slower rate than G1. Although a major fraction of these proteins was eventually transported to the Golgi complex, they did not exhibit as clearly defined a pattern of accumulation as G1, but rather appeared to be distributed throughout the endoplasmic reticulum as well as the Golgi complex. The proteins expressed from constructs lacking most of the cytoplasmic domain and, in some cases, part of the transmembrane domain sequences as well were transported to the cell surface. We have also constructed chimeric proteins with the envelope protein of a murine leukemia virus (MCFenv), which is efficiently transported to the plasma membrane. A MCF-G1 chimera that contained the G1 transmembrane and cytoplasmic domains was found to be efficiently retained in the Golgi complex, and a construct that contained only the G1 transmembrane domain was also partially retained in the Golgi complex. Thus, the transmembrane domain as well as a portion of the cytoplasmic domain adjacent to the transmembrane domain are apparently crucial for Golgi retention of the G1 protein.


Assuntos
Complexo de Golgi/metabolismo , Glicoproteínas de Membrana/metabolismo , Phlebovirus/metabolismo , Animais , Sequência de Bases , Chlorocebus aethiops , Clonagem Molecular , Primers do DNA , Desoxirribonuclease HindIII , Retículo Endoplasmático/metabolismo , Expressão Gênica , Células HeLa , Humanos , Glicoproteínas de Membrana/biossíntese , Dados de Sequência Molecular , Plasmídeos , Reação em Cadeia da Polimerase/métodos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Mapeamento por Restrição , Deleção de Sequência , Transdução de Sinais , Transfecção , Células Vero
16.
J Virol ; 66(7): 4233-41, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1534850

RESUMO

The small RNA segment (S segment) of Uukuniemi (UUK) virus encodes two proteins, the nucleocapsid protein (N) and a nonstructural protein (NSs), by an ambisense strategy. The function of NSs has not been elucidated for any of the bunyaviruses expressing this protein. We have now expressed the N and NSs proteins in Sf9 insect cells by using the baculovirus expression system. High yields of both proteins were obtained. A monospecific antibody was raised against gel-purified NSs and used to study the synthesis and localization of the protein in UUK virus-infected BHK21 cells. While the N protein was detected as early as 4 h postinfection (p.i.), NSs was identified only after 8 h p.i. Both proteins were still synthesized at high levels at 24 h p.i. The half-life of NSs was about 1.5 h, while that of the N protein was several hours. Sucrose gradient fractionation of [35S]methionine-labeled detergent-solubilized extracts of infected BHK21 cells indicated that NSs was firmly associated with the 40S ribosomal subunit. This association took place shortly after translation and was partially resistant to 1 M NaCl. NSs expressed by using the T7 vaccinia virus expression system, as well as in vitro-translated NSs, was also associated with the 40S subunit. In contrast, in vitro-translated N protein was found on top of the gradient. Immunolocalization of NSs, in UUK virus-infected cells, by using an affinity-purified antibody showed a granular cytoplasmic staining. A very similar pattern was seen for cells expressing NSs from a cDNA copy by using a vaccinia virus expression system. No staining was observed in the nuclei in either case. Furthermore, NSs was found neither in virions nor in nucleocapsids isolated from infected cells. In vivo labeling with 32Pi indicated that NSs is not phosphorylated. The possible function of NSs is discussed in light of these results.


Assuntos
Capsídeo/metabolismo , Phlebovirus/metabolismo , Proteínas Ribossômicas/metabolismo , Proteínas do Core Viral/metabolismo , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Baculoviridae/genética , Capsídeo/genética , Capsídeo/imunologia , Linhagem Celular , Imunofluorescência , Meia-Vida , Cinética , Phlebovirus/genética , Testes de Precipitina , Proteínas Recombinantes/metabolismo , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Proteínas não Estruturais Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA