Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Arch Virol ; 165(6): 1515, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32206917

RESUMO

Unfortunately, one of the affiliations of author "A. E. Gorbalenya" was missed in original version. The affiliation is updated here.

2.
Arch Virol ; 165(3): 793-797, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31980941

RESUMO

Enteroviruses (EVs) and rhinoviruses (RVs) are significant pathogens of humans and are the subject of intensive clinical and epidemiological research and public health measures, notably in the eradication of poliovirus and in the investigation and control of emerging pathogenic EV types worldwide. EVs and RVs are highly diverse in their antigenic properties, tissue tropism, disease associations and evolutionary relationships, but the latter often conflict with previously developed biologically defined terms, such as "coxsackieviruses", "polioviruses" and "echoviruses", which were used before their genetic interrelationships were understood. This has created widespread formatting problems and inconsistencies in the nomenclature for EV and RV types and species in the literature and public databases. As members of the International Committee for Taxonomy of Viruses (ICTV) Picornaviridae Study Group, we describe the correct use of taxon names for these viruses and have produced a series of recommendations for the nomenclature of EV and RV types and their abbreviations. We believe their adoption will promote greater clarity and consistency in the terminology used in the scientific and medical literature. The recommendations will additionally provide a useful reference guide for journals, other publications and public databases seeking to use standardised terms for the growing multitude of enteroviruses and rhinoviruses described worldwide.


Assuntos
Enterovirus/classificação , Rhinovirus/classificação , Terminologia como Assunto , Humanos
3.
J Gen Virol ; 98(10): 2421-2422, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28884666

RESUMO

The family Picornaviridae comprises small non-enveloped viruses with RNA genomes of 6.7 to 10.1 kb, and contains >30 genera and >75 species. Most of the known picornaviruses infect mammals and birds, but some have also been detected in reptiles, amphibians and fish. Many picornaviruses are important human and veterinary pathogens and may cause diseases of the central nervous system, heart, liver, skin, gastrointestinal tract or upper respiratory tract. Most picornaviruses are transmitted by the faecal-oral or respiratory routes. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the taxonomy of the Picornaviridae, which is available at www.ictv.global/report/picornaviridae.


Assuntos
Infecções por Picornaviridae/transmissão , Infecções por Picornaviridae/veterinária , Picornaviridae/classificação , Picornaviridae/genética , Anfíbios/virologia , Animais , Aves/virologia , Peixes/virologia , Humanos , Mamíferos/virologia , Infecções por Picornaviridae/virologia , Répteis/virologia , Replicação Viral
4.
J Gen Virol ; 92(Pt 11): 2549-2557, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21752966

RESUMO

HeLa cells are used to study the life cycles of many different viruses, including the human rhinoviruses (HRV) in the family Picornaviridae. Although the natural targets of HRV are human bronchial epithelial cells (hBE), it is generally more difficult to obtain and maintain the relevant primary cell cultures, relative to HeLa cells. Given that the HRV are now identified as a major cause of human asthma exacerbations, it becomes important to document how much of the virus biology learned from HeLa cells is common also to natural primary cells. When compared directly in matched infections using A01a virus, the kinetics of RNA replication, the synthesis and processing of viral proteins and the general subcellular localization of key non-structural proteins were resembled in hBE and HeLa cells. Viral-induced shutoff of host cell processes (e.g. nucleo-cytoplasmic trafficking) was also comparable.


Assuntos
Células Epiteliais/virologia , Rhinovirus/fisiologia , Células Cultivadas , Humanos , Rhinovirus/crescimento & desenvolvimento , Cultura de Vírus , Replicação Viral
5.
Arch Virol ; 151(10): 1933-46, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16732496

RESUMO

Interferons induced by viral infections can have powerful immuno- modulatory effects, and several epidemiologic studies have found an association between certain viral infections and reduced prevalence of allergy. We hypothesized that allergenic proteins could be synthesized by a replicating virus, and this construct could be useful as an immunomodulator. To test this hypothesis, we cloned an allergenic protein (ovomucoid [OVM]) into a murine picornavirus (Mengo virus) vector. This plasmid has a multicloning site surrounded by auto-catalytic sequences so that a foreign protein will be cleaved from viral proteins during replication. OVM sequences were cloned in the context of full-length viral genome cDNA, T7 RNA transcripts of this plasmid were transfected into HeLa cells, and recombinant virus plaques appeared on the second passage. Sequence analysis of recombinant viruses derived from individual plaques demonstrated that three viral isolates contained up to 2/3 of the OVM coding sequence, which was retained by the viruses after 5 additional passages in HeLa cells. The experiments verify the stable expression of immunoreactive OVM subunits by replicating viruses. These virus/allergen constructs could provide a tool to evaluate whether intracellular presentation of allergenic proteins in the context of a viral infection could prevent allergic sensitization upon re-challenge.


Assuntos
Alérgenos/biossíntese , Mengovirus/fisiologia , Ovomucina/biossíntese , Vírus Reordenados/fisiologia , Alérgenos/genética , Animais , Infecções por Cardiovirus/virologia , Vetores Genéticos/metabolismo , Vetores Genéticos/fisiologia , Genoma Viral/genética , Células HeLa/metabolismo , Humanos , Mengovirus/genética , Mengovirus/metabolismo , Camundongos , Ovomucina/genética , Ovomucina/imunologia , Plasmídeos/genética , Vírus Reordenados/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia , Recombinação Genética , Inoculações Seriadas , Especificidade da Espécie , Transfecção , Replicação Viral
6.
J Gen Virol ; 85(Pt 10): 2969-2979, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15448360

RESUMO

Human rhinovirus (HRV) 3C protease (3Cpro) plays several important roles in the virus replication cycle. This enzyme cleaves the viral polyprotein at discrete sites to produce mature viral proteins and also inhibits cellular RNA transcription. It is not clear, however, whether the observed transcriptional shutoff activities are due to 3Cpro itself or to 3Cpro-containing precursors, and where 3Cpro exerts its effects within infected cells. To address these questions HeLa cells were infected with HRV-16, stained with polyclonal antibodies directed against 3Cpro and then analysed by laser confocal microscopy. Proteins containing 3Cpro accumulated in nuclei 2-4 h post-infection, and progressively increased in the cytoplasm. Analyses of subcellular extracts demonstrated that 3CD', a minor component among 3Cpro precursors, gave rise to the earliest 3Cpro nuclear signals. Mature 3Cpro and another 3Cpro precursor, 3CD, were also detected in the nucleus, cytoplasm and perinuclear membrane fractions 4 h post-infection. Transfecting cells with 3Cpro, 3CD precursor and 3CD(Delta371) (with deletion of 371 aa at the carboxyl terminus of 3D) demonstrated that the nucleolar localization signal was near the amino terminus of 3D. In addition, 3Cpro precursors were found to co-localize in nuclei with the transcription factor OCT-1 and the nucleolar chaperone B23. Finally, it was demonstrated that HRV-16 3Cpro, 3CD and 3CD(Delta371) could cleave OCT-1. Collectively, these findings suggest that HRV 3CD' and/or 3CD are specifically localized to the nucleoli of infected cells during the early stage of infection, and contribute to the inhibition of cellular RNA transcription via a proteolytic mechanism.


Assuntos
Transporte Ativo do Núcleo Celular , Núcleo Celular/enzimologia , Cisteína Endopeptidases/metabolismo , Precursores Enzimáticos/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Sequência de Aminoácidos , Fracionamento Celular , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Fator 1 de Transcrição de Octâmero , Fatores de Transcrição/metabolismo
7.
Virology ; 312(2): 481-94, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12919752

RESUMO

Short poly(C)-tract Mengoviruses have proven vaccine efficacy in many species of animals. A novel vector for the delivery of foreign proteins was created by insertion of a second autoproteolytic primary cleavage cassette linked to a multiple cloning site (MCS) into an attenuated variant of Mengo. Nineteen cDNAs from foreign sequences that ranged from 39 to 1653 bases were cloned into the MCS. The viral reading frame was maintained and translation resulted in dual, autocatalytic excision of the foreign peptides without disruption of any Mengo proteins. All cDNAs except those with the largest insertions produced viable virus. Active proteins such as GFP, CAT, and SIV p27 were expressed within infected cells. Relative to parental Mengo, the growth kinetics and genetic stability of each vector was inversely proportional to the size of the inserted sequence. While segments up to 1000 bases could be carried, inserts greater than 500-600 bases were usually reduced in size during serial passage. The limit on carrying capacity was probably due to difficulties in virion assembly or particle stability. Yet for inserts less than 500-600 bases, the Mengo vectors provided an effective system for the delivery of foreign epitopes into cells and mice.


Assuntos
Engenharia Genética , Vetores Genéticos/genética , Mengovirus/genética , Mengovirus/patogenicidade , Animais , DNA Complementar/genética , DNA Viral/genética , Expressão Gênica , Genoma Viral , Células HeLa , Humanos , Mengovirus/fisiologia , Camundongos , Ensaio de Placa Viral , Virulência
8.
J Virol ; 75(23): 11621-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11689644

RESUMO

Recent advances toward using pig tissues in human transplantation have made it necessary to determine the risk of transmitting zoonotic viruses from pigs to humans or vice versa. We investigated the suitability of the porcine encephalomyocarditis virus (EMCV) model for such studies by determining its ability to persist in pigs, escape detection by routine serological methods, and infect human cells. Intraperitoneal inoculation of 5-week-old pigs with EMCV-30, a strain isolated from commercial pigs, resulted in acute cellular degeneration, infiltration of lymphocytes, and apoptosis in myocardium in 13 of 15 (86.7%) pigs during the acute phase of disease (3 to 21 days postinfection), followed by less-severe lymphocytic infiltration and apoptosis in 5 of 10 (50%) pigs during the chronic phase of the disease (day 45 to 90 postinfection). In the brain, lymphocytic infiltration, neuronal degeneration, and gliosis were observed in 26 to 33% of pigs in the acute phase of disease whereas perivascular cuffing was the predominant feature during chronic disease. EMCV antigens and RNA were demonstrated in the myocardium and brain during the chronic phase of disease. Analysis of 100 commercial pigs that were negative for EMCV antibodies identified two pig hearts positive for EMCV RNA. Porcine EMCV productively infected primary human cardiomyocytes as demonstrated by immunostaining using a monoclonal antibody specific for EMCV RNA polymerase, which is expressed only in productively infected cells, and by a one-step growth curve that showed production of 100 to 1,000 PFU of virus per cell within 6 h. The findings that porcine EMCV can persist in pig myocardium and can infect human myocardial cells make it an important infectious agent to screen for in pig-to-human cardiac transplants and a good model for xenozoonosis.


Assuntos
Encefalomielite Enzoótica Suína/virologia , Enterovirus/patogenicidade , Coração/virologia , Animais , Antígenos Virais/análise , Sequência de Bases , Células Cultivadas , Primers do DNA , Enterovirus/genética , Enterovirus/imunologia , Enterovirus/isolamento & purificação , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Modelos Animais , Miocárdio/citologia , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos
9.
Biol Reprod ; 65(6): 1648-54, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11717124

RESUMO

The effect of low concentrations of inorganic phosphate (P(i)) on development, metabolic activity, and mitochondrial organization in the same cohorts of cultured hamster embryos was evaluated. Two-cell embryos were collected from eCG-stimulated golden hamsters and cultured in HECM-10 with 0.0 (control), 1.25, 2.5, or 5.0 microM KH(2)PO(4). Glucose utilization through the Embden-Meyerhof pathway (EMP) and tricarboxylic acid (TCA)-cycle activity were determined following 5 h of culture. Mitochondrial organization in living embryos was evaluated using multiphoton microscopy at 6 h of culture. Development was assessed at 27 h (on-time 8-cell stage) and 51 h (on-time blastocyst stage) of culture. Total cell numbers, as well as cell allocation to the trophectoderm and inner cell mass were determined for morula- and blastocyst-stage embryos. Culture with P(i) did not alter TCA-cycle activity. However, culture with > or 2.5 microM P(i) significantly increased (P < 0.01) EMP activity compared to control. Mitochondrial organization was significantly (P < 0.01) disrupted by P(i) in a dose-dependent manner. Development to the 8-cell, morula/blastocyst, and blastocyst stages was significantly reduced (P < 0.05) in the presence of > or =2.5 microM P(i) compared to both control and 1.25 microM P(i). This study clearly demonstrates that, for hamster embryos, inclusion of even exceptionally low concentrations of P(i) in culture medium dramatically alters embryo physiology. Additionally, although 2-cell embryos can tolerate some structural disruption without concomitant, detrimental effects on development or metabolic activity, metabolic disturbance is associated with decreased developmental competence.


Assuntos
Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fosfatos/administração & dosagem , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/fisiologia , Gonadotropina Coriônica/farmacologia , Ciclo do Ácido Cítrico , Cricetinae , Meios de Cultura , Técnicas de Cultura , Embrião de Mamíferos/ultraestrutura , Glucose/metabolismo , Concentração de Íons de Hidrogênio , Mitocôndrias/ultraestrutura , Mórula/efeitos dos fármacos , Mórula/fisiologia , Concentração Osmolar , Fosfatos/farmacologia
10.
J Virol ; 75(15): 7215-8, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11435606

RESUMO

The cotranslational, primary self-cleavage reaction of cardiovirus polyprotein relies on a highly conserved, short segment of amino acids at the 2A-2B protein boundary. The amino terminus of the required element for encephalomyocarditis virus has now been mapped to include Tyr(126) of the 2A protein, the 18th amino acid before the cleavage site.


Assuntos
Cisteína Endopeptidases/metabolismo , Vírus da Encefalomiocardite/enzimologia , Proteínas Virais , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Cisteína Endopeptidases/genética , DNA Viral , Vírus da Encefalomiocardite/genética , Dados de Sequência Molecular , Mutagênese
11.
J Virol ; 75(7): 3111-20, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11238838

RESUMO

An alignment of cardiovirus sequences led to the prediction of three conserved stem-loops in the 3' untranslated region (UTR) of mengovirus. Deletions of each stem were engineered in mengovirus cDNAs and also in mengovirus replicons, in which part of the viral capsid sequences were replaced with the firefly luciferase gene. The effect of deletion on RNA infectivity and plaque phenotype was evaluated after transfection of viral transcripts into HeLa cells or by luciferase assays of cellular extracts after transfection with RNA replicons. Stem I (mengovirus bases 7666 to 7687) was found to be dispensable for viral growth or exponential luciferase expression. Deletion of stem III (bases 7711 to 7721) was lethal to the virus, and the replicons were incapable of RNA synthesis. Deletion of stem II (DeltaII; bases 7692 to 7705) produced an intermediate phenotype, in that replicons had marginal RNA synthesis activity but transfection with genomic RNA usually failed to produce plaques after normal incubation times (31 h, 37 degrees C). In a few of the DeltaII transfections, however, plaques were observed after long incubation, especially if the cells received large amounts of RNA (3 microg per 3 x 10(6) cells). Viruses from two DeltaII-derived plaques were isolated and amplified. Their RNAs were converted into cDNA, sequenced, and mapped for genotype. Each maintained the DeltaII deletion and, in addition, had one or two reversion mutations, which were characterized by reverse genetics as responsible for the phenotypes. One reversion caused an amino acid change in the polymerase (3D(pol)), and the other was localized to the 3' UTR, upstream of stem I.


Assuntos
Regiões 3' não Traduzidas/química , Mengovirus/genética , Animais , Sequência de Bases , Feminino , Células HeLa , Humanos , Mengovirus/patogenicidade , Mengovirus/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Fenótipo , Filogenia , RNA Viral/biossíntese
12.
Virology ; 290(2): 261-71, 2001 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-11883190

RESUMO

Encephalomyocarditis virus (EMCV) is the prototype member of the cardiovirus genus of picornaviruses. For cardioviruses and the related aphthoviruses, the first protein segment translated from the plus-strand RNA genome is the Leader protein. The aphthovirus Leader (173-201 amino acids) is an autocatalytic papain-like protease that cleaves translation factor eIF-4G to shut off cap-dependent host protein synthesis during infection. The less characterized cardioviral Leader is a shorter protein (67-76 amino acids) and does not contain recognizable proteolytic motifs. Instead, these Leaders have sequences consistent with N-terminal zinc-binding motifs, centrally located tyrosine kinase phosphorylation sites, and C-terminal, acid-rich domains. Deletion mutations, removing the zinc motif, the acid domain, or both domains, were engineered into EMCV cDNAs. In all cases, the mutations gave rise to viable viruses, but the plaque phenotypes in HeLa cells were significantly smaller than for wild-type virus. RNA transcripts containing the Leader deletions had reduced capacity to direct protein synthesis in cell-free extracts and the products with deletions in the acid-rich domains were less effective substrates at the L/P1 site, for viral proteinase 3Cpro. Recombinant EMCV Leader (rL) was expressed in bacteria and purified to homogeneity. This protein bound zinc stoichiometrically, whereas protein with a deletion in the zinc motif was inactive. Polyclonal mouse sera, raised against rL, immunoprecipitated Leader-containing precursors from infected HeLa cell extracts, but did not detect significant pools of the mature Leader. However, additional reactions with antiphosphotyrosine antibodies show that the mature Leader, but not its precursors, is phosphorylated during viral infection. The data suggest the natural Leader may play a role in regulation of viral genome translation, perhaps through a triggering phosphorylation event.


Assuntos
Vírus da Encefalomiocardite/metabolismo , Poliproteínas/metabolismo , Biossíntese de Proteínas , Proteínas Virais/metabolismo , Zinco/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sistema Livre de Células , Vírus da Encefalomiocardite/crescimento & desenvolvimento , Genoma Viral , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Fosforilação , Poliproteínas/genética , Recombinação Genética , Proteínas Virais/genética
13.
Viral Immunol ; 13(1): 27-35, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10733166

RESUMO

We have shown that genetically engineered mengoviruses containing artificially shortened 5' noncoding poly(C) tracts (e.g., C0 or C13UC10) are dramatically attenuated in adult Swiss/ICR mice when compared to wild-type virus or to a genetically engineered virus containing a wild-type length poly(C) tract (C44UC10). To explore further the relationship between poly(C) tracts and virulence, we have conducted more extensive characterizations of several engineered viruses in the murine model. Both short and long poly(C) tract viruses were highly virulent in newborn mice, underscoring the importance of age in poly(C)-mediated attenuation. Virus vMC24, with a tract sequence of C13UC10, was as attenuated in 4-week-old BALB/c, C.C3-H2k/LiMcdJ, and DBA/2 mice as in Swiss/ICR mice. But it was more pathogenic for C57BL/6 mice, and highly virulent for C3H/Hej and C3H/Hen mice, demonstrating the importance of murine genotype. As expected from its virulence in all mouse strains, vMwt, with a poly(C) of C44UC10, induced higher levels of viremia than vMC24. The vMwt also induced higher levels of circulating interferon and had reduced pathogenicity in chemically immunosuppressed Swiss/ICR mice. Similar immunosuppression did not increase the virulence of vMC24. Collectively, the data suggest that endogenous immune components and the immune competence of the host play significant roles in determining the susceptibility of mice to mengovirus infection.


Assuntos
Infecções por Cardiovirus/virologia , Engenharia Genética , Mengovirus/genética , Mengovirus/patogenicidade , Animais , Animais Recém-Nascidos , Encéfalo/virologia , Feminino , Terapia de Imunossupressão , Interferons/sangue , Dose Letal Mediana , Mengovirus/fisiologia , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Viremia/virologia , Virulência , Replicação Viral
14.
J Virol ; 74(7): 3074-81, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10708422

RESUMO

Many virulent aphthoviruses and cardioviruses have long homopolymeric poly(C) tracts in the 5' untranslated regions of their RNA genomes. A panel of genetically engineered mengo-type cardioviruses has been described which contain a variety of different poly(C) tract lengths. Studies of these viruses have shown the poly(C) tract to be dispensable for growth in HeLa cells, although the relative murine virulence of the viruses correlates directly and positively with tract length. Compared with wild-type mengovirus strain M, mutants with shortened poly(C) tracts grow poorly in mice and protectively immunize rather than kill recipient animals. In the present study, several murine cell populations were tested to determine whether, unlike HeLa cells, they allowed a differential amplification of viruses with long or short poly(C) tracts. Replication and cytopathic studies with four hematopoietically derived cell lines (CH2B, RAW 264.7, A20.J, and P815) and two murine fibroblast cell lines [L929 and L(Y)] demonstrated that several of these cell types indeed allowed differential virus replication as a function of viral poly(C) tract length. Among the most discerning of these cells, RAW 264.7 macrophages supported vigorous lytic growth of a long-tract virus, vMwt (C(44)UC(10)), but supported only substantially diminished and virtually nonlytic growth of vMC(24) (C(13)UC(10)) and vMC(0) short-tract viruses. The viral growth differences evident in all cell lines were apparent early and continuously during every cycle of virus amplification. The data suggest that poly(C) tract-dependent attenuation of mengovirus may be due in part to a viral replication defect manifest in similar hematopoietic-type cells shortly after murine infection. The characterized cultures should provide excellent tools for molecular study of poly(C) tract-mediated virulence.


Assuntos
Vírus da Encefalomiocardite/crescimento & desenvolvimento , Mengovirus/crescimento & desenvolvimento , Poli C/genética , Animais , Linhagem Celular , Efeito Citopatogênico Viral , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/fisiologia , Células HeLa , Humanos , Interferons/fisiologia , Mengovirus/genética , Mengovirus/fisiologia , Camundongos
15.
Virology ; 260(1): 148-55, 1999 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-10405366

RESUMO

Measurement of an antigenic response to the aphthovirus infection-associated antigen (VIA), the viral RNA polymerase 3D(pol), is frequently used as a discriminating assay for the extent of viral replication in animals. In practice, animals seropositive for VIA are assumed to have been exposed to live virus, although in fact it is suspected that endogenous 3D(pol) in commercial inactivated vaccines may occasionally stimulate analogous responses and result in false-positive tests for virus exposure. Cardiovirus infections in mice produce similar anti-VIA antibodies, and in view of recently developed attenuated Mengo vaccines and live Mengo vectors, these VIA responses are also under investigation as potential correlates of vaccine efficacy. We have purified recombinant Mengo 3D(pol), developed monoclonal antibodies to the protein, and used these reagents in highly sensitive Western blot assays to quantify the levels of endogenous 3D(pol) in Mengo and encephalomyocarditis virus (EMCV) preparations. The presence of 3D(pol) was detected at all stages of standard vaccine purification procedures, including materials purified by CsCl. Clarified suspensions of Mengo- or encephalomyocarditis virus-infected HeLa cells were found to contain very high quantities of 3D(pol), averaging approximately 1.2-1.5 micrograms of protein/micrograms of virus. Pelleting through 30% sucrose or purification by CsCl removed much of this material, but even these samples retained approximately 0.2-0.4 ng of 3D(pol)/micrograms virus. These ratios represent approximately 1 3D(pol) molecule/20 virus particles in the most highly purified materials and probably indicate that 3D(pol) is a contaminant on the particle surface rather than an intrinsically packaged molecule. In clarified cell lysates, which are commonly used as vaccine inocula, the protein to virus ratio was approximately 210:1, a level that could represent serious contamination problems for future VIA detection if such inocula are used without further purification.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Vírus da Encefalomiocardite/enzimologia , Mengovirus/enzimologia , RNA Polimerase Dependente de RNA , Animais , Western Blotting , Centrifugação com Gradiente de Concentração , Efeito Citopatogênico Viral , Eletroforese em Gel de Poliacrilamida , Células HeLa , Humanos , Camundongos , Cultura de Vírus
16.
J Wildl Dis ; 35(2): 384-7, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10231768

RESUMO

Encephalomyocarditis virus (EMCV), has caused the deaths of many species of animals in zoological parks and research institutions. The Audubon Park Zoo, (New Orleans, Louisiana, USA) attempted vaccination of several species with a killed EMCV vaccine with mixed results. This paper reports an attempt at vaccination against EMCV using a genetically engineered, live attenuated Mengo virus (vMC0) at the Audubon Park Zoo and Miami Metro Zoo, (Miami, Florida, USA) from December 1996 to June 1997. Several species of animals were vaccinated with vMC0, which is serologically indistinguishable from the field strain of EMCV. Serum samples were taken at the time of vaccination and again 21 days later, then submitted for serum neutralization titers against EMCV. The vaccinate species included red capped mangebey (Cercocebus torquatus), colobus (Colobus guereza), angolan colobus (Colobus angolensis), ruffed lemur (Lemur variegatus ruber and Lemur variegatus variegatus), back lemur (Lemur macaco), ring-tailed lemur (Lemur catta), siamang (Hylobates syndactylus), diana guenon (Cercopithicus diana), spider monkey (Ateles geoffroyi), common marmoset (Callithrix jacchus), talapoin monkey (Cercopithecus talapoin), Brazilian tapir (Tapirus terrestris), Baird's tapir (Tapirus bairdii), Malayan tapir (Tapirus indicus), dromedary camel (Camelus dromedarius), bactrian camel (Camelus bactrianus), gerenuk (Litocranius walleri), guanaco (Lama glama guanicoe), black duiker (Cephalophus niger), Vietnamese potbellied pig (Sus scrofa), babirusa (Babyrousa babyrussa), collard peccary (Tayass tajacu), and African crested porcupine (Hystrix africaeaustralis). The vaccine response was variable, with high virus neutralizing antibody titer responses in some primate species and mixed to poor responses for other species. No ill effects were seen with vaccination.


Assuntos
Animais de Zoológico , Infecções por Cardiovirus/veterinária , Mengovirus/imunologia , Vacinas Virais , Animais , Artiodáctilos , Infecções por Cardiovirus/prevenção & controle , Engenharia Genética , Células HeLa , Humanos , Mengovirus/genética , Perissodáctilos , Primatas , Roedores , Vacinas Atenuadas/genética , Vacinas Sintéticas/genética , Vacinas Virais/genética
17.
Adv Exp Med Biol ; 440: 759-65, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9782355

RESUMO

Recent studies suggest that infectious viruses and particularly persisting viral RNAs often exist as diverse populations or "quasispecies". We have developed an approach to characterize populations of the murine coronavirus mouse hepatitis virus (MHV) generated during persistent infection which has allowed us to begin to address the role of the viral quasispecies in MHV pathogenesis. We analyzed the population of persisting viral RNAs using reverse-transcription polymerase chain reaction amplification (RT-PCR) of the S1 "hypervariable" region of the spike gene followed by differential colony hybridization to identify spike deletion variants (SDVs) from acute and persistently infected mice. Sequence analysis revealed that mice with the most severe chronic paralysis harbored the most complex quasispecies. Mapping of the SDVs to the predicted RNA secondary structure of the spike RNA revealed that an isolated stem loop structure is frequently deleted. Overall, these results are consistent with high frequency recombination at sites of RNA secondary structure contributing to expansion of the viral quasispecies and persisting viral pathogenesis.


Assuntos
Glicoproteínas de Membrana/genética , Vírus da Hepatite Murina/genética , RNA Viral , Proteínas do Envelope Viral/genética , Latência Viral , Animais , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/fisiologia , Recombinação Genética , Glicoproteína da Espícula de Coronavírus
18.
J Virol ; 72(7): 5811-9, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9621041

RESUMO

Inhibitors of the phosphatidylinositol 3-kinase (PI3 kinase)-FKBP-rapamycin-associated protein (FRAP) pathway, such as rapamycin and wortmannin, induce dephosphorylation and activation of the suppressor of cap-dependent translation, 4E-BP1. Encephalomyocarditis virus (EMCV) infection leads to activation of 4E-BP1 at the time of host translation shutoff. Consistent with these data, rapamycin mildly enhances the synthesis of viral proteins and the shutoff of host cell protein synthesis after EMCV infection. In this study, two defective EMCV strains were generated by deleting portions of the 2A coding region of an infectious cDNA clone. These deletions dramatically decreased the efficiency of viral protein synthesis and abolished the virus-induced shutoff of host translation after infection of BHK-21 cells. Both translation and processing of the P1-2A capsid precursor polypeptide are impaired by the deletions in 2A. The translation and yield of mutant viruses were increased significantly by the presence of rapamycin and wortmannin during infection. Thus, inhibition of the PI3 kinase-FRAP signaling pathway partly complements mutations in 2A protein and reverses a slow-virus phenotype.


Assuntos
Androstadienos/farmacologia , Vírus Defeituosos/efeitos dos fármacos , Vírus da Encefalomiocardite/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Polienos/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Células Cultivadas , Cricetinae , Vírus da Encefalomiocardite/fisiologia , Biossíntese de Proteínas , Coelhos , Sirolimo , Proteínas Virais/biossíntese , Wortmanina
19.
J Virol ; 71(8): 6183-90, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9223514

RESUMO

Coronavirus RNA evolves in the central nervous systems (CNS) of mice during persistent infection. This evolution can be monitored by detection of a viral quasispecies of spike deletion variants (SDVs) (C. L. Rowe, S. C. Baker, M. J. Nathan, and J. O. Fleming, J. Virol. 71:2959-2969, 1997). We and others have found that the deletions cluster in the region from 1,200 to 1,800 nucleotides from the 5' end of the spike gene sequence, termed the "hypervariable" region. To address how SDVs might arise, we generated the predicted folding structures of the positive- and negative-strand senses of the entire 4,139-nt spike RNA sequence. We found that a prominent, isolated stem-loop structure is coincident with the hypervariable region in each structure. To determine if this predicted stem-loop is a "hot spot" for RNA recombination, we assessed whether this region of the spike is more frequently deleted than three other selected regions of the spike sequence in a population of viral sequences isolated from the CNS of acutely and persistently infected mice. Using differential colony hybridization of cloned spike reverse transcription-PCR products, we detected SDVs in which the hot spot was deleted but did not detect SDVs in which other regions of the spike sequence were exclusively deleted. Furthermore, sequence analysis and mapping of the crossover sites of 25 distinct patterns of SDVs showed that the majority of crossover sites clustered to two regions at the base of the isolated stem-loop, which we designated as high-frequency recombination sites 1 and 2. Interestingly, the majority of the left and right crossover sites of the SDVs were directly across from or proximal to one another, suggesting that these SDVs are likely generated by intramolecular recombination. Overall, our results are consistent with there being an important role for the spike RNA secondary structure as a contributing factor in the generation of SDVs during persistent infection.


Assuntos
DNA Viral/química , Deleção de Genes , Vírus da Hepatite Murina/genética , Recombinação Genética , Sequência de Bases , Dados de Sequência Molecular
20.
J Virol ; 70(11): 8182-6, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8892950

RESUMO

The RNA genomes from the cardioviruses, hepatoviruses, and aphthoviruses encode two to five tandem pseudoknots within their 5' untranslated regions. These pseudoknots lie adjacent to a pyrimidine-rich sequence, which in cardio- and aphthoviruses takes the form of a homopolymeric poly(C) tract. Seven deletion mutations within mengovirus pseudoknots PK(B) and PK(C) were created and characterized. tested in tissue culture, mengovirus genomes with alterations in PK(C) were viable but had small plaque phenotypes. Larger plaque revertants were isolated and partially characterized, and each proved to be a second-site pseudorevertant with (unmapped) changes elsewhere in the genome. The infectious PK(C) mutant viruses were highly lethal to mice, and deletions in this motif did not affect mengovirus virulence in the same manner as deletions in the adjacent poly(C) tract. In contrast, deletions in PK(B), or deletions which spanned PK(B) + PK(C), produced nonviable genomes. Cell-free translations directed by any of the altered PK sequences gave normal polyprotein amounts relative to wild-type mengovirus. But viral RNA accumulation during HeLa cell infection was dramatically impaired, even with the least disruptive of the PK(C) changes, suggesting the pseudoknots play an essential though undefined role in RNA synthesis and moreover that an intact PK(B) structure is critical to this function.


Assuntos
Mengovirus/genética , Poli C/metabolismo , RNA Viral/biossíntese , Animais , Células HeLa , Humanos , Mengovirus/patogenicidade , Camundongos , Camundongos Endogâmicos ICR , Deleção de Sequência , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA