Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 93(12)2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30944180

RESUMO

Equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) represent two members of the family Arteriviridae and pose major threats for the horse- and swine-breeding industries worldwide. A previous study suggested that PRRSV nsp4, a 3C-like protease, antagonizes interferon beta (IFN-ß) production by cleaving the NF-κB essential modulator (NEMO) at a single site, glutamate 349 (E349). Here, we demonstrated that EAV nsp4 also inhibited virus-induced IFN-ß production by targeting NEMO for proteolytic cleavage and that the scission occurred at four sites: E166, E171, glutamine 205 (Q205), and E349. Additionally, we found that, besides the previously reported cleavage site E349 in NEMO, scission by PRRSV nsp4 took place at two additional sites, E166 and E171. These results imply that while cleaving NEMO is a common strategy utilized by EAV and PRRSV nsp4 to antagonize IFN induction, EAV nsp4 adopts a more complex substrate recognition mechanism to target NEMO. By analyzing the abilities of the eight different NEMO fragments resulting from EAV or PRRSV nsp4 scission to induce IFN-ß production, we serendipitously found that a NEMO fragment (residues 1 to 349) could activate IFN-ß transcription more robustly than full-length NEMO, whereas all other NEMO cleavage products were abrogated for the IFN-ß-inducing capacity. Thus, NEMO cleavage at E349 alone may not be sufficient to completely inactivate the IFN response via this signaling adaptor. Altogether, our findings suggest that EAV and PRRSV nsp4 cleave NEMO at multiple sites and that this strategy is critical for disarming the innate immune response for viral survival.IMPORTANCE The arterivirus nsp4-encoded 3C-like protease (3CLpro) plays an important role in virus replication and immune evasion, making it an attractive target for antiviral therapeutics. Previous work suggested that PRRSV nsp4 suppresses type I IFN production by cleaving NEMO at a single site. In contrast, the present study demonstrates that both EAV and PRRSV nsp4 cleave NEMO at multiple sites and that this strategy is essential for disruption of type I IFN production. Moreover, we reveal that EAV nsp4 also cleaves NEMO at glutamine 205 (Q205), which is not targeted by PRRSV nsp4. Notably, targeting a glutamine in NEMO for cleavage has been observed only with picornavirus 3C proteases (3Cpro) and coronavirus 3CLpro In aggregate, our work expands knowledge of the innate immune evasion mechanisms associated with NEMO cleavage by arterivirus nsp4 and describes a novel substrate recognition characteristic of EAV nsp4.


Assuntos
Equartevirus/metabolismo , Interferon beta/biossíntese , Proteínas não Estruturais Virais/metabolismo , Animais , Arteriviridae/metabolismo , Arterivirus/metabolismo , Linhagem Celular , Equartevirus/fisiologia , Células HEK293 , Cavalos , Humanos , Quinase I-kappa B/metabolismo , Quinase I-kappa B/fisiologia , Evasão da Resposta Imune , Imunidade Inata , Interferon beta/metabolismo , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Proteólise , Transdução de Sinais , Suínos , Replicação Viral
2.
J Biol Chem ; 293(31): 12054-12067, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-29887523

RESUMO

Nidovirus endoribonucleases (NendoUs) include nonstructural protein 15 (nsp15) from coronaviruses and nsp11 from arteriviruses, both of which have been reported to participate in the viral replication process and in the evasion of the host immune system. Results from a previous study of coronaviruses SARS-CoV, HCoV-229E, and MHV nsp15 indicate that it mainly forms a functional hexamer, whereas nsp11 from the arterivirus PRRSV is a dimer. Here, we found that porcine Deltacoronavirus (PDCoV) nsp15 primarily exists as dimers and monomers in vitro Biological experiments reveal that a PDCoV nsp15 mutant lacking the first 27 amino acids of the N-terminal domain (Asn-1-Asn-27) forms more monomers and displays decreased enzymatic activity, indicating that this region is important for its dimerization. Moreover, multiple sequence alignments and three-dimensional structural analysis indicated that the C-terminal region (His-251-Val-261) of PDCoV nsp15 is 10 amino acids shorter and forms a shorter loop than that formed by the equivalent sequence (Gln-259-Phe-279) of SARS-CoV nsp15. This result may explain why PDCoV nsp15 failed to form hexamers. We speculate that NendoUs may have originated from XendoU endoribonucleases (XendoUs) forming monomers in eukaryotic cells, that NendoU from arterivirus gained the ability to form dimers, and that the coronavirus variants then evolved the capacity to assemble into hexamers. We further propose that PDCoV nsp15 may be an intermediate in this evolutionary process. Our findings provide a theoretical basis for improving our understanding of NendoU evolution and offer useful clues for designing drugs and vaccines against nidoviruses.


Assuntos
Coronavirus/química , Endorribonucleases/química , Nidovirales/química , Subunidades Proteicas/química , Proteínas não Estruturais Virais/química , Sequência de Aminoácidos , Arterivirus/química , Arterivirus/classificação , Arterivirus/genética , Arterivirus/metabolismo , Sítios de Ligação , Clonagem Molecular , Coronavirus/classificação , Coronavirus/genética , Coronavirus/metabolismo , Cristalografia por Raios X , Endorribonucleases/genética , Endorribonucleases/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Evolução Molecular , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Modelos Moleculares , Nidovirales/classificação , Nidovirales/genética , Nidovirales/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/genética
3.
Virus Res ; 246: 28-34, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29337162

RESUMO

Recently, a novel antiviral compound (K22) that inhibits replication of a broad range of animal and human coronaviruses was reported to interfere with viral RNA synthesis by impairing double-membrane vesicle (DMV) formation (Lundin et al., 2014). Here we assessed potential antiviral activities of K22 against a range of viruses representing two (sub)families of the order Nidovirales, the Arteriviridae (porcine reproductive and respiratory syndrome virus [PRRSV], equine arteritis virus [EAV] and simian hemorrhagic fever virus [SHFV]), and the Torovirinae (equine torovirus [EToV] and White Bream virus [WBV]). Possible effects of K22 on nidovirus replication were studied in suitable cell lines. K22 concentrations significantly decreasing infectious titres of the viruses included in this study ranged from 25 to 50 µM. Reduction of double-stranded RNA intermediates of viral replication in nidovirus-infected cells treated with K22 confirmed the anti-viral potential of K22. Collectively, the data show that K22 has antiviral activity against diverse lineages of nidoviruses, suggesting that the inhibitor targets a critical and conserved step during nidovirus replication.


Assuntos
Antivirais/farmacologia , Arterivirus/efeitos dos fármacos , Benzamidas/farmacologia , Coronaviridae/efeitos dos fármacos , Equartevirus/efeitos dos fármacos , Piperidinas/farmacologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/efeitos dos fármacos , Torovirus/efeitos dos fármacos , Animais , Arterivirus/genética , Arterivirus/crescimento & desenvolvimento , Arterivirus/metabolismo , Carpas , Linhagem Celular , Chlorocebus aethiops , Coronaviridae/genética , Coronaviridae/crescimento & desenvolvimento , Coronaviridae/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Equartevirus/genética , Equartevirus/crescimento & desenvolvimento , Equartevirus/metabolismo , Mesocricetus , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/crescimento & desenvolvimento , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , RNA de Cadeia Dupla/antagonistas & inibidores , RNA de Cadeia Dupla/biossíntese , RNA de Cadeia Dupla/genética , RNA Viral/antagonistas & inibidores , RNA Viral/biossíntese , RNA Viral/genética , Torovirus/genética , Torovirus/crescimento & desenvolvimento , Torovirus/metabolismo , Replicação Viral/efeitos dos fármacos
4.
Virus Res ; 220: 70-90, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27071852

RESUMO

All eukaryotic positive-stranded RNA (+RNA) viruses appropriate host cell membranes and transform them into replication organelles, specialized micro-environments that are thought to support viral RNA synthesis. Arteriviruses (order Nidovirales) belong to the subset of +RNA viruses that induce double-membrane vesicles (DMVs), similar to the structures induced by e.g. coronaviruses, picornaviruses and hepatitis C virus. In the last years, electron tomography has revealed substantial differences between the structures induced by these different virus groups. Arterivirus-induced DMVs appear to be closed compartments that are continuous with endoplasmic reticulum membranes, thus forming an extensive reticulovesicular network (RVN) of intriguing complexity. This RVN is remarkably similar to that described for the distantly related coronaviruses (also order Nidovirales) and sets them apart from other DMV-inducing viruses analysed to date. We review here the current knowledge and open questions on arterivirus replication organelles and discuss them in the light of the latest studies on other DMV-inducing viruses, particularly coronaviruses. Using the equine arteritis virus (EAV) model system and electron tomography, we present new data regarding the biogenesis of arterivirus-induced DMVs and uncover numerous putative intermediates in DMV formation. We generated cell lines that can be induced to express specific EAV replicase proteins and showed that DMVs induced by the transmembrane proteins nsp2 and nsp3 form an RVN and are comparable in topology and architecture to those formed during viral infection. Co-expression of the third EAV transmembrane protein (nsp5), expressed as part of a self-cleaving polypeptide that mimics viral polyprotein processing in infected cells, led to the formation of DMVs whose size was more homogenous and closer to what is observed upon EAV infection, suggesting a regulatory role for nsp5 in modulating membrane curvature and DMV formation.


Assuntos
Arterivirus/ultraestrutura , Membrana Celular/ultraestrutura , Retículo Endoplasmático/ultraestrutura , Organelas/ultraestrutura , Organelas/virologia , Proteínas não Estruturais Virais/genética , Animais , Arterivirus/genética , Arterivirus/metabolismo , Infecções por Arterivirus/veterinária , Infecções por Arterivirus/virologia , Linhagem Celular , Membrana Celular/virologia , Coronavirus/genética , Coronavirus/metabolismo , Coronavirus/ultraestrutura , Tomografia com Microscopia Eletrônica , Retículo Endoplasmático/virologia , Expressão Gênica , Interações Hospedeiro-Patógeno , Proteínas não Estruturais Virais/metabolismo
5.
DNA Cell Biol ; 34(12): 728-35, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26398903

RESUMO

NLRP3 inflammasome, which is multiprotein complex that induces the maturity and secretion of proinflammatory interleukin-1ß (IL-1ß), takes a bridge between the innate and adaptive immune responses to the invading pathogens. It has been shown that porcine reproductive and respiratory syndrome virus (PRRSV) could activate the NLRP3 inflammasome but induce the host's immunosuppression. This study aims to explore whether PRRSV could encode the component to antagonize the NLRP3 inflammasome. The obtained results showed that PRRSV could induce the expression and secretion of IL-1ß in early infection through the pathway of NLRP3 inflammasome in porcine alveolar macrophages (PAMs), but the levels of pro-IL-1ß mRNA and IL-1ß protein decreased to a degree that was similar to the level of the mock-infected group in later infection. This work also found that PRRSV nonstructural protein (nsp) 11 could inhibit the expression of pro-IL-1ß mRNA induced by lipopolysaccharide (LPS) and the secretion of IL-1ß induced by LPS plus nigericin in PAMs. Furthermore, the mutation studies showed that the endoribonuclease activity was essential for nsp11 to inhibit the secretion of IL-1ß. Therefore, it could be indicated that PRRSV could induce the activation of NLRP3 inflammasome, but the virus encoded nsp11 to inhibit this action.


Assuntos
Arterivirus/fisiologia , Proteínas de Transporte/metabolismo , Endorribonucleases/metabolismo , Inflamassomos/metabolismo , Interleucina-1beta/biossíntese , Síndrome Respiratória e Reprodutiva Suína/virologia , Proteínas não Estruturais Virais/metabolismo , Animais , Arterivirus/metabolismo , Interleucina-1beta/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA Mensageiro/genética , Suínos
6.
J Virol ; 88(16): 9129-40, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24899184

RESUMO

UNLABELLED: The N-terminal region of simian hemorrhagic fever virus (SHFV) nonstructural polyprotein 1a is predicted to encode three papain-like proteases (PLP1α, PLP1ß, and PLP1γ). Catalytic residues and cleavage sites for each of the SHFV PLP1s were predicted by alignment of the SHFV PLP1 region sequences with each other as well as with those of other arteriviruses, and the predicted catalytic residues were shown to be proximal by homology modeling of the SHFV nsp1s on porcine respiratory and reproductive syndrome virus (PRRSV) nsp1 crystal structures. The functionality of the predicted catalytic Cys residues and cleavage sites was tested by analysis of the autoproteolytic products generated in in vitro transcription/translation reactions done with wild-type or mutant SHFV nsp1 constructs. Cleavage sites were also analyzed by mass spectroscopy analysis of selected immunoprecipitated cleavage products. The data showed that each of the three SHFV PLP1s is an active protease. Cys63 was identified as the catalytic Cys of SHFV PLP1α and is adjacent to an Ala instead of the canonical Tyr observed in other arterivirus PLP1s. SHFV PLP1γ is able to cleave at both downstream and upstream nsp1 junction sites. Although intermediate precursor polyproteins as well as alternative products generated by each of the SHFV PLP1s cleaving at sites within the N-terminal region of nsp1ß were produced in the in vitro reactions, Western blotting of SHFV-infected, MA104 cell lysates with SHFV nsp1 protein-specific antibodies detected only the three mature nsp1 proteins. IMPORTANCE: SHFV is unique among arteriviruses in having three N-terminal papain-like protease 1 (PLP1) domains. Other arteriviruses encode one or two active PLP1s. This is the first functional study of the SHFV PLP1s. Analysis of the products of in vitro autoprocessing of an N-terminal SHFV nonstructural 1a polypeptide fragment showed that each of the three SHFV PLP1s is active, and the predicted catalytic Cys residues and cleavage sites for each PLP1 were confirmed by testing mutant constructs. Several unique features of the SHFV PLP1s were discovered. The SHFV PLP1α catalytic Cys63 is unique among arterivirus PLP1s in being adjacent to an Ala instead of a Trp. Other arterivirus PLP1s cleave only in cis at a single downstream site, but SHFV PLP1γ can cleave at both the downstream nsp1γ-nsp2 and upstream nsp1ß-nsp1γ junctions. The three mature nsp1 proteins were produced both in the in vitro reactions and in infected cells.


Assuntos
Arterivirus/genética , Papaína/genética , Papaína/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Animais , Arterivirus/metabolismo , Infecções por Arterivirus/virologia , Catálise , Linhagem Celular , Dados de Sequência Molecular , Poliproteínas/genética , Poliproteínas/metabolismo , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Alinhamento de Sequência , Suínos/virologia
7.
Proc Natl Acad Sci U S A ; 109(43): E2920-8, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23043113

RESUMO

Programmed -1 ribosomal frameshifting (-1 PRF) is a gene-expression mechanism used to express many viral and some cellular genes. In contrast, efficient natural utilization of -2 PRF has not been demonstrated previously in eukaryotic systems. Like all nidoviruses, members of the Arteriviridae (a family of positive-stranded RNA viruses) express their replicase polyproteins pp1a and pp1ab from two long ORFs (1a and 1b), where synthesis of pp1ab depends on -1 PRF. These polyproteins are posttranslationally cleaved into at least 13 functional nonstructural proteins. Here we report that porcine reproductive and respiratory syndrome virus (PRRSV), and apparently most other arteriviruses, use an additional PRF mechanism to access a conserved alternative ORF that overlaps the nsp2-encoding region of ORF1a in the +1 frame. We show here that this ORF is translated via -2 PRF at a conserved G_GUU_UUU sequence (underscores separate ORF1a codons) at an estimated efficiency of around 20%, yielding a transframe fusion (nsp2TF) with the N-terminal two thirds of nsp2. Expression of nsp2TF in PRRSV-infected cells was verified using specific Abs, and the site and direction of frameshifting were determined via mass spectrometric analysis of nsp2TF. Further, mutagenesis showed that the frameshift site and an unusual frameshift-stimulatory element (a conserved CCCANCUCC motif 11 nucleotides downstream) are required to direct efficient -2 PRF. Mutations preventing nsp2TF expression impair PRRSV replication and produce a small-plaque phenotype. Our findings demonstrate that -2 PRF is a functional gene-expression mechanism in eukaryotes and add another layer to the complexity of arterivirus genome expression.


Assuntos
Arterivirus/metabolismo , Mutação da Fase de Leitura , Ribossomos/metabolismo , Proteínas Virais/biossíntese , Sequência de Aminoácidos , Animais , Sequência de Bases , Compartimento Celular , Códon , Mamíferos , Espectrometria de Massas , Dados de Sequência Molecular , Mutagênese , Fases de Leitura Aberta , Homologia de Sequência do Ácido Nucleico , Proteínas Virais/genética
8.
J Biol Chem ; 283(24): 16525-36, 2008 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-18411274

RESUMO

The cytoplasmic replication of positive-stranded RNA viruses is associated with characteristic, virus-induced membrane structures that are derived from host cell organelles. We used the prototype arterivirus, equine arteritis virus (EAV), to gain insight into the structure and function of the replication/transcription complex (RTC) of nidoviruses. RTCs were isolated from EAV-infected cells, and their activity was studied using a newly developed in vitro assay for viral RNA synthesis, which reproduced the synthesis of both viral genome and subgenomic mRNAs. A detailed characterization of this system and its reaction products is described. RTCs isolated from cytoplasmic extracts by differential centrifugation were inactive unless supplemented with a cytosolic host protein factor, which, according to subsequent size fractionation analysis, has a molecular mass in the range of 59-70 kDa. This host factor was found to be present in a wide variety of eukaryotes. Several EAV replicase subunits cosedimented with newly made viral RNA in a heavy membrane fraction that contained all RNA-dependent RNA polymerase activity. This fraction contained the characteristic double membrane vesicles (DMVs) that were previously implicated in EAV RNA synthesis and could be immunolabeled for EAV nonstructural proteins (nsps). Replicase subunits directly involved in viral RNA synthesis (nsp9 and nsp10) or DMV formation (nsp2 and nsp3) exclusively cosedimented with the active RTC. Subgenomic mRNAs appeared to be released from the complex, whereas newly made genomic RNA remained more tightly associated. Taken together, our data strongly support a link between DMVs and the RNA-synthesizing machinery of arteriviruses.


Assuntos
Arterivirus/metabolismo , Equartevirus/metabolismo , Fatores Hospedeiros de Integração/metabolismo , Animais , Membrana Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Citoplasma/metabolismo , Células HeLa , Humanos , Cloreto de Lítio/farmacologia , Modelos Biológicos , RNA Viral/metabolismo , Frações Subcelulares/metabolismo , Células Vero
9.
Virol J ; 2: 29, 2005 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-15823209

RESUMO

Although PCR and RT-PCR provided a valuable approach for detection of pathogens, the high level of sensitivity of these assays also makes them prone to false positive results. In addition to cross-contamination with true positive samples, false positive results are also possible due to "carry-over" contamination of samples with amplicon DNA generated by previous reactions. To reduce this source of false positives, amplicon generated by reactions in which dUTP was substituted for dTTP can be degraded by uracil DNA glycosylase (UNG). UNG does not degrade RNA but will cleave contaminating uracil-containing DNA while leaving thymine-containing DNA intact. The availability of heat-labile UNG makes use of this approach feasible for RT-PCR. In this study, real-time RT-PCR was used to quantify UNG degradation of amplicon DNA and the effect of UNG on RNA detection. Using the manufacturers' recommended conditions, complete degradation of DNA was not observed for samples containing 250 copies of amplicon DNA. Doubling the UNG concentration resulted in degradation of the two lowest concentrations of DNA tested, but also resulted in an increase of 1.94 cycles in the CT for RNA detection. To improve DNA degradation while minimizing the effect on RNA detection, a series of time, temperature and enzyme concentrations were evaluated. Optimal conditions were found to be 0.25 U UNG per 25 microl reaction with a 20 min, 30 degrees C incubation prior to RT-PCR. Under these conditions, high concentrations of amplicon DNA could be degraded while the CT for RNA detection was increased by 1.2 cycles.


Assuntos
DNA Viral/análise , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Uracila-DNA Glicosidase/metabolismo , Arterivirus/isolamento & purificação , Arterivirus/metabolismo , DNA Viral/metabolismo , Reações Falso-Positivas , RNA Viral/metabolismo , Sensibilidade e Especificidade
14.
J Virol ; 72(5): 4341-51, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9557724

RESUMO

The 3' noncoding region (NCR) of the negative-strand RNA [3'(-)NCR RNA] of the arterivirus simian hemorrhagic fever virus (SHFV) is 209 nucleotides (nt) in length. Since this 3' region, designated 3'(-)209, is the site of initiation of full-length positive-strand RNA and is the template for the synthesis of the 5' leader sequence, which is found on both full-length and subgenomic mRNAs, it is likely to contain cis-acting signals for RNA synthesis and to interact with cellular and viral proteins to form replication complexes. Gel mobility shift assays showed that cellular proteins in MA104 S100 cytoplasmic extracts formed two complexes with the SHFV 3'(-)209 RNA, and results from competition gel mobility shift assays demonstrated that these interactions were specific. Four proteins with molecular masses of 103, 86, 55, and 36 kDa were detected in UV-induced cross-linking assays, and three of these proteins (103, 55, and 36 kDa) were also detected by Northwestern blotting assays. Identical gel mobility shift and UV-induced cross-linking patterns were obtained with uninfected and SHFV-infected extracts, indicating that the four proteins detected are cellular, not viral, proteins. The binding sites for the four cellular proteins were mapped to the region between nt 117 and 184 (68-nt sequence) from the 3' end of the SHFV negative-strand RNA. This 68-nt sequence was predicted to form two stem-loops, SL4 and SL5. The 3'(-)NCR RNA of another arterivirus, lactate dehydrogenase-elevating virus C (LDV-C), competed with the SHFV 3'(-)209 RNA in competition gel mobility shift assays. UV-induced cross-linking assays showed that four MA104 cellular proteins with the same molecular masses as those that bind to the SHFV 3'(-)209 RNA also bind to the LDV-C 3'(-)NCR RNA and equine arteritis virus 3'(-)NCR RNA. However, each of these viral RNAs also bound to an additional MA104 protein. The binding sites for the MA104 cellular proteins were shown to be located in similar positions in the LDV-C 3'(-)NCR and SHFV 3'(-)209 RNAs. These data suggest that the binding sites for a set of the cellular proteins are conserved in all arterivirus RNAs and that these cell proteins may be utilized as components of viral replication complexes.


Assuntos
Arterivirus/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Arterivirus/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Sequência Conservada , Flavivirus/genética , Macaca mulatta , Dados de Sequência Molecular , Peso Molecular , Conformação de Ácido Nucleico , RNA Viral/química , Proteínas de Ligação a RNA/química
15.
Virology ; 221(1): 98-112, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8661418

RESUMO

Porcine Reproductive and Respiratory Syndrome Virus (PRRSV), a small enveloped virus containing a positive-strand RNA genome, possesses at least three major structural proteins designated N, M, and E. The N protein is considered as the major component of the nucleocapsid, whereas M and E are membrane-associated. Previous studies using peptide-specific antibodies assigned these proteins to ORFs 7, 6, and 5, respectively. In the present report, monospecific antisera raised against Escherichia coli-expressed ORFs 5, 6, and 7 products were used to study the synthesis and processing of PRRSV structural proteins in the highly permissive MARC-145 cell line. Treatment of viral proteins with various glycosidases showed that only E was modified by N-linked glycans. Pulse-chase experiments revealed that intracellular transport of the major envelope glycoprotein was delayed in the premedial Golgi compartment. During the first 30 min of chase, E undergoes a gradual downward shift of its apparent molecular weight, thought to result from trimming of the mannose-rich glycan structures. Once E is transported to the medial Golgi or proximal elements, some molecules undergo complete processing of all their high-mannose N-linked oligosaccharides to complex type, while in other molecules only a fraction of N-linked glycans are terminally glycosylated. These two differentially glycosylated forms of E were found to be incorporated into extracellular virions. In cells and virions, both M and E were shown to occur in heterodimeric complexes linked by disulfide bonds. The oligomerization process, as analyzed from pulse-chase experiments, showed that M and E are incorporated into M-E complexes with different kinetics and efficiencies, in a fashion similar to their counterparts in equine arteritis virus. Apparently, all steps of E protein N-glycans processing proceed after its association with M which occurs in the endoplasmic reticulum (ER). In the infected cells, E and M appear highly membrane-associated, while N is predominantly cytosolic.


Assuntos
Arterivirus/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Arterivirus/genética , Arterivirus/fisiologia , Sequência de Bases , Transporte Biológico , Linhagem Celular , DNA Viral , Glicosilação , Cinética , Manose/metabolismo , Dados de Sequência Molecular , Oligossacarídeos/metabolismo , Fases de Leitura Aberta , Coelhos , Proteínas do Envelope Viral , Proteínas Estruturais Virais/genética , Vírion/metabolismo , Montagem de Vírus
16.
J Gen Virol ; 77 ( Pt 6): 1265-70, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8683215

RESUMO

The subgenomic mRNAs (sg mRNA) of porcine reproductive and respiratory syndrome virus (PRRSV) were characterized. The number of sg mRNAs, which form a 3'-coterminal nested set in PRRSV-infected cells, varied from six to seven among PRRSV isolates with differing virulence. The additional species of sg mRNA in some isolates of PRRSV was designated as sg mRNA 3-1. The leader-mRNA junction sequences of sg mRNAs 3, 3-1 and 4 were found to contain a similar six nucleotide sequence motif, U(G)UA(G/C)ACC. By comparing the 5'-terminal sequence of sg mRNA 3-1 with the genomic sequence of two isolates, ISU79 and ISU1894, it was found that a point mutation, from U in isolate ISU1894 to C in isolate ISU79, led to the acquisition of a new leader-mRNA junction sequence (UUGACC) in isolate ISU79, and therefore an additional species of sg mRNA 3-1. A small ORF (3-1) was identified at the 5' end of sg mRNA 3-1.


Assuntos
Infecções por Arterivirus/veterinária , Arterivirus/genética , RNA Mensageiro/biossíntese , Doenças dos Suínos , Sequência de Aminoácidos , Animais , Arterivirus/isolamento & purificação , Arterivirus/metabolismo , Infecções por Arterivirus/virologia , Sequência de Bases , Northern Blotting , Linhagem Celular , Códon , Primers do DNA , Sondas de DNA , DNA Complementar , Dados de Sequência Molecular , Fases de Leitura Aberta , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Suínos , Regiões Terminadoras Genéticas
17.
Virology ; 206(1): 155-63, 1995 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-7831770

RESUMO

The genome of Lelystad virus (LV), a positive-strand RNA virus, is 15 kb in length and contains 8 open reading frames (ORFs) that encode putative viral proteins. ORFs 2 to 7 were cloned in plasmids downstream of the Sp6 RNA polymerase promoter, and the translation of transcripts generated in vitro yielded proteins that could be immunoprecipitated with porcine anti-LV serum. Synthetic polypeptides of 15 to 17 amino acids were selected from the amino acid sequences of ORFs 2 to 7 and antipeptide sera were raised in rabbits. Antisera that immunoprecipitated the in vitro translation products of ORFs 2 to 5 and 7 were obtained. Sera containing antibodies directed against peptides from ORFs 3 to 7 reacted positively with LV-infected alveolar lung macrophages in the immunoperoxidase monolayer assay. Using these antipeptide sera and porcine anti-LV serum, we identified three structural proteins and assigned their corresponding genes. Virions were found to contain a nucleocapsid protein of 15 kDa (N), an unglycosylated membrane protein of 18 kDa (M), and a glycosylated membrane protein of 25 kDa (E). The N protein is encoded by ORF7, the M protein is encoded by ORF6, and the E protein is encoded by ORF5. The E protein in virus particles contains one or two N-glycans that are resistant to endo-beta-N-acetyl-D-glucosaminidase H. This finding indicates that the high-mannose glycans are processed into complex glycans in the Golgi compartment. The protein composition of the LV virions further confirms that LV is evolutionarily related to equine arteritis virus, simian hemorrhagic fever virus, and lactate dehydrogenase-elevating virus.


Assuntos
Arterivirus/metabolismo , Proteínas Estruturais Virais/genética , Sequência de Aminoácidos , Animais , Arterivirus/genética , Sequência de Bases , Células Cultivadas , Glicosídeo Hidrolases/metabolismo , Soros Imunes , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Fases de Leitura Aberta , Fragmentos de Peptídeos/imunologia , Testes de Precipitina , Biossíntese de Proteínas , Suínos , Transcrição Gênica , Proteínas Estruturais Virais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...