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








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 7(2): e32061, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22355409

RESUMO

Infections with the picornavirus, human rhinovirus (HRV), are a major cause of wheezing illnesses and asthma exacerbations. In developing a murine model of picornaviral airway infection, we noted the absence of murine rhinoviruses and that mice are not natural hosts for HRV. The picornavirus, mengovirus, induces lethal systemic infections in its natural murine hosts, but small genetic differences can profoundly affect picornaviral tropism and virulence. We demonstrate that inhalation of a genetically attenuated mengovirus, vMC(0), induces lower respiratory tract infections in mice. After intranasal vMC(0) inoculation, lung viral titers increased, peaking at 24 h postinoculation with viral shedding persisting for 5 days, whereas HRV-A01a lung viral titers decreased and were undetectable 24 h after intranasal inoculation. Inhalation of vMC(0), but not vehicle or UV-inactivated vMC(0), induced an acute respiratory illness, with body weight loss and lower airway inflammation, characterized by increased numbers of airway neutrophils and lymphocytes and elevated pulmonary expression of neutrophil chemoattractant CXCR2 ligands (CXCL1, CXCL2, CXCL5) and interleukin-17A. Mice inoculated with vMC(0), compared with those inoculated with vehicle or UV-inactivated vMC(0), exhibited increased pulmonary expression of interferon (IFN-α, IFN-ß, IFN-λ), viral RNA sensors [toll-like receptor (TLR)3, TLR7, nucleotide-binding oligomerization domain containing 2 (NOD2)], and chemokines associated with HRV infection in humans (CXCL10, CCL2). Inhalation of vMC(0), but not vehicle or UV-inactivated vMC(0), was accompanied by increased airway fluid myeloperoxidase levels, an indicator of neutrophil activation, increased MUC5B gene expression, and lung edema, a sign of infection-related lung injury. Consistent with experimental HRV inoculations of nonallergic, nonasthmatic human subjects, there were no effects on airway hyperresponsiveness after inhalation of vMC(0) by healthy mice. This novel murine model of picornaviral airway infection and inflammation should be useful for defining mechanisms of HRV pathogenesis in humans.


Assuntos
Mengovirus/genética , Mengovirus/patogenicidade , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Animais , Western Blotting , Modelos Animais de Doenças , Edema/imunologia , Edema/metabolismo , Edema/virologia , Feminino , Expressão Gênica , Humanos , Interferons/metabolismo , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Linfócitos/imunologia , Linfócitos/metabolismo , Linfócitos/virologia , Mengovirus/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/virologia , Infecções por Picornaviridae/imunologia , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/virologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Infecções Respiratórias/imunologia , Eliminação de Partículas Virais/genética , Redução de Peso
2.
J Gen Virol ; 91(Pt 5): 1239-44, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20089798

RESUMO

Both entero- and cardioviruses have been shown to suppress host mRNA synthesis. Enteroviruses are also known to inhibit the activity of rRNA genes, whereas this ability of cardioviruses is under debate. This study reported that mengovirus (a cardiovirus) suppressed rRNA synthesis but less efficiently than poliovirus (an enterovirus). In contrast to poliovirus infection, the incorporation of BrUTP, fluorouridine and [14C]uridine in rRNA precursors was observed even during the late stages of mengovirus infection, although at a significantly reduced level. The cleavage of TATA-binding protein, considered to be one of the central events in poliovirus-induced transcription shutoff, was not detected in mengovirus-infected cells, indicating a difference in the mechanisms of host RNA synthesis inhibition caused by these viruses. The results also showed that functional leader protein is redundant for the suppression of host RNA synthesis by cardiovirus.


Assuntos
Interações Hospedeiro-Patógeno , Mengovirus/patogenicidade , Poliovirus/patogenicidade , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Ribossômico/antagonistas & inibidores , RNA Ribossômico/biossíntese , Células HeLa , Humanos
3.
Cell Microbiol ; 12(3): 310-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19863558

RESUMO

Picornaviruses encompass a large family of RNA viruses. Some picornaviruses possess a leader (L) protein at the N-terminus of their polyprotein. The L proteins of encephalomyocarditis virus, a cardiovirus, and foot-and-mouth disease virus (FMDV), an aphthovirus, are both dispensable for replication and their major function seems to be the suppression of antiviral host cell responses. Previously, we showed that the L protein of mengovirus, a strain of encephalomyocarditis virus, inhibits antiviral responses by inhibiting type I interferon (IFN-alpha/beta) gene transcription. The L protein of the FMDV is a protease (L(pro)) that cleaves cellular factors to reduce cytokine and chemokine mRNA production and to inhibit cap-dependent cellular host mRNA translation, thereby limiting the production of proteins with antiviral activity. In this study, we constructed a viable chimeric mengovirus that expresses FMDV L(pro) in place of the authentic L protein in order to compare the efficiency of the immune evasion mechanisms mediated by L and L(pro) respectively. We show that in this mengovirus background the L protein is more potent than FMDV L(pro) in suppressing IFN-alpha/beta responses. Yet, FMDV L(pro) is important to antagonize infection-limiting responses both in vitro and in vivo.


Assuntos
Vírus da Febre Aftosa/imunologia , Vírus da Febre Aftosa/patogenicidade , Interferon-alfa/imunologia , Interferon beta/imunologia , Mengovirus/imunologia , Mengovirus/patogenicidade , Proteínas Virais/imunologia , Animais , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/virologia , Linhagem Celular , Cricetinae , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Interferon-alfa/antagonistas & inibidores , Interferon beta/antagonistas & inibidores , Mengovirus/genética , Mengovirus/crescimento & desenvolvimento , Camundongos , Recombinação Genética , Análise de Sobrevida , Carga Viral , Proteínas Virais/genética , Fatores de Virulência/genética , Fatores de Virulência/imunologia
4.
Virol J ; 6: 122, 2009 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-19671179

RESUMO

BACKGROUND: Infection of the lower airways by rhinovirus, a member of the picornavirus family, is an important cause of wheezing illnesses in infants, and plays an important role in the pathogenesis of rhinovirus-induced asthma exacerbations. Given the absence of natural rhinovirus infections in rodents, we investigated whether an attenuated form of mengovirus, a picornavirus whose wild-type form causes systemic rather than respiratory infections in its natural rodent hosts, could induce airway infections in rats with inflammatory responses similar to those in human rhinovirus infections. RESULTS: After inoculation with 10(7) plaque-forming units of attenuated mengovirus through an inhalation route, infectious mengovirus was consistently recovered on days 1 and 3 postinoculation from left lung homogenates (median Log10 plaque-forming units = 6.0 and 4.8, respectively) and right lung bronchoalveolar lavage fluid (median Log10 plaque-forming units = 5.8 and 4.0, respectively). Insufflation of attenuated mengovirus, but not vehicle or UV-inactivated virus, into the lungs of BN rats caused significant increases (P < 0.05) in lower airway neutrophils and lymphocytes in the bronchoalveolar lavage fluid and patchy peribronchiolar, perivascular, and alveolar cellular infiltrates in lung tissue sections. In addition, infection with attenuated mengovirus significantly increased (P < 0.05) lower airway levels of neutrophil chemoattractant CXCR2 ligands [cytokine-induced neutrophil chemoattractant-1 (CINC-1; CXCL1) and macrophage inflammatory protein-2 (MIP-2; CXCL2)] and monocyte chemoattractant protein-1 (MCP-1; CCL2) in comparison to inoculation with vehicle or UV-inactivated virus. CONCLUSION: Attenuated mengovirus caused a respiratory infection in rats with several days of viral shedding accompanied by a lower airway inflammatory response consisting of neutrophils and lymphocytes. These features suggest that mengovirus-induced airway infection in rodents could be a useful model to define mechanisms of rhinovirus-induced airway inflammation in humans.


Assuntos
Modelos Animais de Doenças , Mengovirus/patogenicidade , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Animais , Humanos , Inflamação/patologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Linfócitos/imunologia , Masculino , Mengovirus/imunologia , Neutrófilos/imunologia , Infecções por Picornaviridae/imunologia , Ratos , Infecções Respiratórias/imunologia , Eliminação de Partículas Virais
5.
J Virol ; 77(17): 9136-46, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12915530

RESUMO

Previous studies using wild-type Encephalomyocarditis virus (EMCV) and Mengo virus, which have long poly(C) tracts (61 to 146 C's) at the 5' nontranslated region of the genome, and variants of these viruses genetically engineered to truncate or substitute the poly(C) tracts have produced conflicting data on the role of the poly(C) tract in the virulence of these viruses. Analysis of the nucleotide sequence of an EMCV strain isolated from an aborted swine fetus (EMCV 30/87) revealed that the virus had a poly(C) tract that was 7- to 10-fold shorter than the poly(C) tracts of other EMCV strains and 4-fold shorter than that of Mengo virus. Subsequently, we investigated the virulence and pathogenesis of this naturally occurring short-poly(C)-tract-containing virus in rodents, pigs, and nonhuman primates. Infection of C57BL/6 mice, pigs, and cynomolgus macaques resulted in similar EMCV 30/87 pathogenesis, with the heart and brain as the primary sites of infections in all three animals, but with different disease phenotypes. Sixteen percent of EMCV 30/87-infected pigs developed acute fatal cardiac failure, whereas the rest of the pigs were overtly asymptomatic for as long as 90 days postinfection (p.i.), despite extensive myocardial and central nervous system (CNS) pathological changes. In contrast, mice infected with >/==" BORDER="0">4 PFU of EMCV 30/87 developed acute encephalitis that resulted in the death of all animals (n = 25) between days 2 and 7 p.i. EMCV 30/87-infected macaques remained overtly asymptomatic for 45 days, despite extensive myocardial and CNS pathological changes and viral persistence in more than 50% of the animals. The short poly(C) tract in EMCV 30/87 (CUC(5)UC(8)) was comparable to that of strain 2887A/91 (C(10)UCUC(3)UC(10)), another recent porcine isolate.


Assuntos
Infecções por Cardiovirus/etiologia , Encefalomielite Enzoótica Suína/etiologia , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/patogenicidade , Animais , Sequência de Bases , Encéfalo/patologia , Encéfalo/virologia , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/virologia , Encefalomielite Enzoótica Suína/patologia , Encefalomielite Enzoótica Suína/virologia , Vírus da Encefalomiocardite/classificação , Coração/virologia , Humanos , Macaca fascicularis , Mengovirus/genética , Mengovirus/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Filogenia , Poli C/genética , RNA Viral/genética , Especificidade da Espécie , Sus scrofa , Virulência/genética
6.
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
7.
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
8.
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
9.
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
10.
Virology ; 223(2): 344-50, 1996 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-8806569

RESUMO

We have shown previously that genetically engineered Mengo viruses with artificial deletions in their 5' noncoding polyribocytidylic acid (poly(C)) tracts are highly attenuated for the natural murine host and also for other animals such as baboons, macaques, and domestic pigs. The present report further characterizes select short poly(C) tract Mengo viruses in the natural murine host. A positive correlation was found between the length of the poly(C) tract and murine virulence, as measured by virus brain titers and brain lesion scores after infection. Histological examination of brain tissue collected from infected animals clearly showed that the short poly(C) tract viruses did not induce the devastating pathological effects characteristic of animals inoculated with wild-type virus. Instead, the short-tract Mengo viruses proved excellent immunological agents. A dose of only 100 plaque-forming units of vMC24 (poly(C) tract: C13UC10), injected subcutaneously, protected 80% of recipient animals against a normally lethal dose of encephalomyocarditis virus. The protection was long-lived, and animals similarly immunized with vMCo virus (poly(C) tract: Co) still had protective neutralizing antibody titers up to 16 months after inoculation. In addition, the short-tract viruses proved genetically stable, in that the vMC24 virus did not yield detectable pathogenic revertants even after multiple, forced passages in 4-week-old mice. These studies suggest that Mengo viruses containing deletions in their poly(C) tracts are biologically safe and potent immunogens and imply that they may have uses as cardiovirus vaccines.


Assuntos
Infecções por Cardiovirus/imunologia , Mengovirus/genética , Mengovirus/imunologia , Poli C/genética , Animais , Anticorpos Antivirais/imunologia , Encéfalo/patologia , Encéfalo/virologia , Infecções por Cardiovirus/virologia , Relação Dose-Resposta Imunológica , Feminino , Mengovirus/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Deleção de Sequência , Vacinação , Virulência
11.
J Virol ; 70(3): 2027-31, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8627731

RESUMO

Previously, we described three mengovirus mutants derived from cDNA plasmids, containing shortened poly(C) tracts (C8, C12, and C13UC10), that exhibited strong attenuation for virulence in mice yet grew like wild-type virus in HeLa cells. Thirteen additional mutants hav now been constructed and characterized. Five of these differ only in poly(C) length, including one with a precise deletion of the tract. The other mutants bear deletions into the regions juxtaposing poly(C). Studies with HeLa cells confirm the essential dispensability of mengovirus's poly(C) tract but reveal a subtle, measurable correlation between poly(C) length and plaque diameter. Virulence studies with mice also revealed a strong correlation between poly(C) length and virulence. For the poly(C)-flanking mutations, the 15 bases directly 5' of the tract proved dispensable for virus viability, whereas the 20 to 30 bases 3' of poly(C) were critical for growth, thus implicating this region in the basal replication of the virus.


Assuntos
Mengovirus/genética , Poli C , Animais , Sequência de Bases , Infecções por Cardiovirus/virologia , Clonagem Molecular , Técnicas de Cultura , DNA Complementar , DNA Viral , Feminino , Células HeLa , Humanos , Mengovirus/metabolismo , Mengovirus/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Mutagênese , Conformação de Ácido Nucleico , Poli C/genética , RNA Viral/metabolismo , Virulência/genética , Replicação Viral/genética
12.
J Virol ; 69(4): 2697-9, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7884926

RESUMO

We have constructed three cDNA clones of encephalomyocarditis virus strain R (EMCV-R) with poly(C) tracts of C4, C9, and C20. RNA transcribed from these cDNAs was infectious to HeLa cells, and the resultant viruses grew well in this system, albeit with plaque sizes that were proportional to the poly(C) length. When injected into mice, the progeny viruses were only slightly less pathogenic than EMCV-R, and the observed degree of attenuation was not nearly as dramatic as for equivalent mengoviruses with similar short poly(C)s. Short-tract poly(C)-mediated attenuation is therefore highly dependent on viral genomic context.


Assuntos
Vírus da Encefalomiocardite/patogenicidade , Mengovirus/patogenicidade , Poli C/genética , Animais , Clonagem Molecular , DNA Complementar , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/fisiologia , Feminino , Células HeLa , Humanos , Mengovirus/genética , Mengovirus/fisiologia , Camundongos , Ensaio de Placa Viral , Virulência/genética
13.
Nature ; 343(6257): 474-6, 1990 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-2153940

RESUMO

The murine cardioviruses, such as the Mengo and encephalomyocarditis viruses, and the bovine aphthoviruses, such as foot-and-mouth disease virus, are distinguished among positive-strand RNA viruses by the presence of long homopolymeric poly(C) tracts within their 5' noncoding sequences. Although the specific lengths (60-350 bases) and sequence discontinuities (for example, uridine residues) that sometimes disrupt the homopolymer have served to characterize natural viral isolates, the biological function of the poly(C) region has never been clear. We now report that complementary DNA-mediated truncation of the Mengo virus poly(C) tract dramatically attenuates the pathogenicity of the virus in mice. Animals injected with viruses with short tracts not only survived inoculation of up to 50 micrograms live virus (10(11) plaque-forming units) but consistently produced high titres of neutralizing antibodies, which conferred long-term immunogenic protection from (normally) lethal virus challenge. We propose that analogous synthetic strains of foot and mouth disease virus could serve as the basis for new attenuated vaccines.


Assuntos
Genes Virais , Engenharia Genética , Mengovirus/patogenicidade , Poli C/genética , Polirribonucleotídeos/genética , Animais , Anticorpos Antivirais/análise , Antígenos Virais/imunologia , Clonagem Molecular , DNA/genética , Infecções por Enterovirus/imunologia , Infecções por Enterovirus/prevenção & controle , Mengovirus/genética , Mengovirus/imunologia , Meningoencefalite/etiologia , Camundongos
14.
Arch Virol ; 93(1-2): 31-49, 1987.
Artigo em Inglês | MEDLINE | ID: mdl-3028343

RESUMO

Biological properties of two mengovirus mutants, 205 and 280, were compared to those of wild-type virus. The mutants exhibited alterations in plaque morphology, hemagglutination, and virulence in mice, but were not temperature-sensitive. Agglutination of human erythrocytes by mengovirus was dependent on the presence of sialic acid on the erythrocyte surface; however, free sialic acid failed to inhibit hemagglutination. Glycophorin, the major sialoglycoprotein of human erythrocyte membranes, exhibited receptor specificity for wild-type virus, but not for mutants 205 or 280. Cross-linking studies indicated that glycophorin exhibited binding specificity for the alpha (1 D) structural protein. The LD50 titers for wild-type mengovirus were 7 and 1500 plaque forming units (PFU) in mice infected intracranially (IC) and intraperitoneally (IP), respectively. However, mice infected IC or IP with 10(6) or 10(7) PFU of mutant 205 or 280 did not exhibit symptoms indicative of virus infection. Revertants were isolated from the brains of mice infected with mutant 205, but not from the brains of mice infected with mutant 280. The biological characterization of the revertants indicated that hemagglutination and virulence may be phenotypically-linked traits.


Assuntos
Hemaglutininas Virais/genética , Mengovirus/fisiologia , Animais , Anticorpos Antivirais/imunologia , Cricetinae , Eritrócitos/microbiologia , Glicoforinas/metabolismo , Hemaglutininas Virais/metabolismo , Mengovirus/genética , Mengovirus/imunologia , Mengovirus/patogenicidade , Camundongos , Mutação , Testes de Neutralização , Fenótipo , Ratos/sangue , Receptores Virais/fisiologia , Ovinos/sangue , Temperatura , Replicação Viral
15.
J Virol ; 50(3): 684-90, 1984 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-6327999

RESUMO

In mice, Mengovirus produces a fatal encephalitis. Plaque purification of the virus resulted in the isolation of a clone (Mengo- 2T ), which in addition to encephalitis caused diabetes. Microscopic examination of pancreases from infected mice revealed necrosis in the islets of Langerhans and infiltration of inflammatory cells. By immunofluorescence viral antigens were found in the islets, and radioimmunoassays demonstrated a substantial decrease in pancreatic immunoreactive insulin. Studies on susceptibility among inbred strains of mice showed that whereas the D variant of encephalomyocarditis virus caused diabetes only in SJL/J mice, Mengo- 2T caused diabetes in strains of mice resistant to encephalomyocarditis-induced diabetes (i.e., CBA/J, C3H/HeJ, CE/J, AKR/J, C57BL/6J). The ability of Mengo- 2T to induce diabetes in encephalomyocarditis-resistant mice was found to be due to the greater capacity of Mengo- 2T as compared to the D variant of encephalomyocarditis virus to replicate in and destroy the islets of these animals. Although Mengo- 2T and the D variant of encephalomyocarditis virus are antigenically indistinguishable by hyperimmune sera, our studies show that these viruses have different host ranges and tissue tropisms .


Assuntos
Diabetes Mellitus Experimental/microbiologia , Vírus da Encefalomiocardite/patogenicidade , Mengovirus/patogenicidade , Animais , Glicemia/análise , Encéfalo/microbiologia , Encéfalo/patologia , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Imunofluorescência , Imunidade Inata , Insulina/análise , Camundongos , Camundongos Endogâmicos , Radioimunoensaio , Especificidade da Espécie
16.
Acta Histochem ; 75(1): 47-62, 1984.
Artigo em Alemão | MEDLINE | ID: mdl-6095576

RESUMO

Infection of ABAF hybride mice with MengoM viruses prepared as a suspension of mouse brain tissue (0.1 ml 10 LD50) after a series of more than 90 animal passages produced a severe panencephalitis and pancreatitis 60 h after inoculation. Histological examinations of the brain, pancreas, and heart revealed damage to the central nervous system and the acinar cells of the pancreas, consisting of a degeneration of the specific cells and their organelles, with little involvement of the heart muscle cells detectable with the electron microscope. This was consistent with high virus titers in the tissues in the period immediately following infection. The acute damage of exocrine tissues was without evidence of diabetes-like changes in the islets of Langerhans.


Assuntos
Encéfalo/patologia , Infecções por Enterovirus/patologia , Miocárdio/patologia , Pâncreas/patologia , Animais , Encéfalo/ultraestrutura , Masculino , Mengovirus/patogenicidade , Camundongos , Camundongos Endogâmicos , Microscopia Eletrônica , Miocárdio/ultraestrutura , Organoides/ultraestrutura , Pâncreas/ultraestrutura
17.
Acta Histochem ; 53(1): 97-102, 1975.
Artigo em Alemão | MEDLINE | ID: mdl-171906

RESUMO

The paper presents a photometric method for the determination of virus-induced alterations (cytopathogenic effects) of cells based on nephelometric analysis. These were taken at varying time intervals after infection and with different multiplicity of infection. It was demonstrated that suppression of virus-induced CPE by the known inhibitors could also be followed by turbidity measurements. The in vitro experiments were carried out in the 2 following virus-cell-systems: Pseudorabies virus in chick-embryo cells and Mengo virus in FL-cells, respectively. According to these studies the measurements were extended to other virus strains and inhibitors (antibiotics, chemicals, and antimetabolites). The results obtained confirm the applicability of the photometric method for the screening of antiviral drugs.


Assuntos
Antivirais/farmacologia , Efeito Citopatogênico Viral/efeitos dos fármacos , Herpesviridae/patogenicidade , Herpesvirus Suídeo 1/patogenicidade , Mengovirus/patogenicidade , Amantadina/farmacologia , Animais , Linhagem Celular , Embrião de Galinha , Cicloeximida/farmacologia , Herpesvirus Suídeo 1/efeitos dos fármacos , Técnicas In Vitro , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/patogenicidade , Mengovirus/efeitos dos fármacos , Nefelometria e Turbidimetria , Vírus da Doença de Newcastle/patogenicidade , Sindbis virus/efeitos dos fármacos , Sindbis virus/patogenicidade , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA