Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733647

RESUMO

Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV hemagglutinin (H) and fusion (F) envelope glycoproteins; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad repeat (HR) regions of F can inhibit MV infection at the entry stage. In previous work, we have generated potent MV fusion inhibitors by dimerizing the F-derived peptides and conjugating them to cholesterol. We have shown that prophylactic intranasal administration of our lead fusion inhibitor efficiently protects from MV infection in vivo We show here that peptides tagged with lipophilic moieties self-assemble into nanoparticles until they reach the target cells, where they are integrated into cell membranes. The self-assembly feature enhances biodistribution and the half-life of the peptides, while integration into the target cell membrane increases fusion inhibitor potency. These factors together modulate in vivo efficacy. The results suggest a new framework for developing effective fusion inhibitory peptides. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. We show here that specific biophysical properties regulate the in vivo efficacy of MV F-derived peptides.


Assuntos
Hemaglutininas Virais/imunologia , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/efeitos dos fármacos , Sarampo/prevenção & controle , Nanopartículas/administração & dosagem , Peptídeos/imunologia , Proteínas Virais de Fusão/imunologia , Administração Intranasal , Sequência de Aminoácidos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Colesterol/química , Feminino , Meia-Vida , Hemaglutininas Virais/química , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Sarampo/imunologia , Sarampo/mortalidade , Sarampo/virologia , Vacina contra Sarampo/síntese química , Vírus do Sarampo/química , Vírus do Sarampo/imunologia , Nanopartículas/química , Peptídeos/síntese química , Sigmodontinae , Análise de Sobrevida , Proteínas Virais de Fusão/química , Internalização do Vírus/efeitos dos fármacos
2.
J Virol ; 89(2): 1143-55, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25378493

RESUMO

UNLABELLED: Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV H and the fusion (F) envelope glycoprotein; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad-repeat (HR) regions of F can potently inhibit MV infection at the entry stage. We show here that specific features of H's interaction with its receptors modulate the susceptibility of MV F to peptide fusion inhibitors. A higher concentration of inhibitory peptides is required to inhibit F-mediated fusion when H is engaged to its nectin-4 receptor than when H is engaged to its CD150 receptor. Peptide inhibition of F may be subverted by continued engagement of receptor by H, a finding that highlights the ongoing role of H-receptor interaction after F has been activated and that helps guide the design of more potent inhibitory peptides. Intranasal administration of these peptides results in peptide accumulation in the airway epithelium with minimal systemic levels of peptide and efficiently prevents MV infection in vivo in animal models. The results suggest an antiviral strategy for prophylaxis in vulnerable and/or immunocompromised hosts. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that parenterally delivered fusion-inhibitory peptides protect mice from lethal CNS MV disease. Here we show, using established small-animal models of MV infection, that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. Since the fusion inhibitors are stable at room temperature, this intranasal strategy is feasible even outside health care settings, could be used to protect individuals and communities in case of MV outbreaks, and could complement global efforts to control measles.


Assuntos
Antivirais/administração & dosagem , Vírus do Sarampo/efeitos dos fármacos , Sarampo/prevenção & controle , Oligopeptídeos/administração & dosagem , Proteínas Virais de Fusão/administração & dosagem , Internalização do Vírus/efeitos dos fármacos , Administração Intranasal , Animais , Quimioprevenção/métodos , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sigmodontinae
3.
Vet Pathol ; 47(4): 677-89, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20442421

RESUMO

Human T-lymphotropic virus type-1 (HTLV-1), the first human retrovirus discovered, is the causative agent of adult T-cell leukemia/lymphoma (ATL) and a number of lymphocyte-mediated inflammatory conditions including HTLV-1-associated myelopathy/tropical spastic paraparesis. Development of animal models to study the pathogenesis of HTLV-1-associated diseases has been problematic. Mechanisms of early infection and cell-to-cell transmission can be studied in rabbits and nonhuman primates, but lesion development and reagents are limited in these species. The mouse provides a cost-effective, highly reproducible model in which to study factors related to lymphoma development and the preclinical efficacy of potential therapies against ATL. The ability to manipulate transgenic mice has provided important insight into viral genes responsible for lymphocyte transformation. Expansion of various strains of immunodeficient mice has accelerated the testing of drugs and targeted therapy against ATL. This review compares various mouse models to illustrate recent advances in the understanding of HTLV-1-associated ATL development and how improvements in these models are critical to the future development of targeted therapies against this aggressive T-cell lymphoma.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Imunidade Inata/imunologia , Leucemia-Linfoma de Células T do Adulto/imunologia , Paraparesia Espástica Tropical/imunologia , Animais , Modelos Animais de Doenças , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Leucemia-Linfoma de Células T do Adulto/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Paraparesia Espástica Tropical/genética
4.
Vet Pathol ; 46(5): 1003-14, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19429977

RESUMO

Infection with human T-cell leukemia virus type 1 (HTLV-1) leads sometimes to the development of adult T-cell lymphoma/leukemia (ATL), which is invariably fatal and often associated with humoral hypercalcemia of malignancy. The transformation of infected CD4 T cells and the pathogenesis of leukemia have been studied with great limitation in tissue culture and patients. To better understand the pathogenesis and perform preclinical drug studies, animal models of ATL are urgently needed. In mice, inoculation of HTLV-1 cell lines mostly leads to development of localized lymphomas. To develop an ATL animal model with leukemic spread of ATL cells, mouse strains with different well-defined immune deficiencies were inoculated intraperitoneally with different HTLV-1-infected cell lines (ACH.2, C8166, MT-2, MET-1). Inoculation of MET-1 cells into NOD/SCID mice provided the best model system for slowly developing T-cell leukemia with multiple organ involvement. In leukemic mice, an increase in serum calcium levels correlated with expression of receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells ligand on leukemic cells and secretion of parathyroid hormone-related protein and interleukin-6. In contrast to the other cell lines that did not spread systemically, MET-1 expressed both the adhesion molecules CD11a (LFA-1alpha) and CD49d (VLA-4alpha) and produced or induced expression of matrix metalloproteinases 1, 2, 3, and 9, thus underlining the importance of these molecules in the spread of adult T-cell leukemia cells. The MET-1/NOD/SCID model will be useful for developing interventions against invasion and spread of leukemic cells and subsequent humoral hypercalcemia of malignancy.


Assuntos
Modelos Animais de Doenças , Hipercalcemia/etiologia , Leucemia-Linfoma de Células T do Adulto/imunologia , Camundongos Endogâmicos NOD/imunologia , Camundongos SCID/imunologia , Animais , Basigina/imunologia , Western Blotting/veterinária , Antígeno CD11a/imunologia , Linhagem Celular , Feminino , Humanos , Hipercalcemia/imunologia , Imuno-Histoquímica/veterinária , Integrina alfa4/imunologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Camundongos Knockout , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/imunologia , Ligante RANK/imunologia , RNA/química , RNA/genética , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária
5.
Clin Exp Immunol ; 148(2): 218-29, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17335559

RESUMO

Epidemiological studies show an association between early exposure to respiratory syncytial virus (RSV) and the development or exacerbation of asthma. This idea is supported by studies in mice that demonstrate worsened airway hyper-reactivity (AHR) when RSV-infected animals are exposed to allergen. The effect of allergen on RSV disease, however, has not been reported. Cotton rats (Sigmodon hispidus) that have been used as a model to study RSV pathogenesis were sensitized to extracts of Aspergillus fumigatus (Af), a common household mould. The allergic response to Af included eosinophilia, formation of granulomas and induction of Th2 type cytokines. RSV infection prior to allergen challenge resulted in exacerbation of the inflammatory response as well as increased airway responsiveness to methacholine. The exacerbated response was indeed dependent on virus replication. Virus replication in turn was influenced by the allergic response, with persistence in the noses for 2 days longer in animals challenged with allergen. This diminished clearance corresponded to decreased induction of mRNA for IFN-gamma, a Th1-type cytokine that is characteristic of viral infection. Treatment of RSV-infected Af-challenged animals with recombinant IFN-gamma reduced the allergic inflammatory response as well as the relative levels of Th1 and Th2 cytokine mRNA. However, this treatment did not reduce airway reactivity, showing that these pathologic and physiologic measures of exacerbated disease are independent. We speculate that the reciprocal effect of the allergic response on viral immunity may benefit the host by limiting exacerbation of physiologic responses that are IFN-gamma-dependent.


Assuntos
Alérgenos/imunologia , Hipersensibilidade Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/fisiologia , Infecções Respiratórias/virologia , Animais , Antivirais/uso terapêutico , Aspergillus fumigatus/imunologia , Citocinas/biossíntese , Citocinas/genética , Feminino , Granuloma/imunologia , Granuloma/patologia , Granuloma/virologia , Interferon gama/uso terapêutico , Masculino , RNA Mensageiro/genética , Proteínas Recombinantes , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/patologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/imunologia , Infecções Respiratórias/patologia , Sigmodontinae , Células Th2/imunologia , Replicação Viral/imunologia
6.
Nat Med ; 7(6): 725-31, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11385511

RESUMO

Surface-contact-mediated signaling induced by the measles virus (MV) fusion and hemagglutinin glycoproteins is necessary and sufficient to induce T-cell unresponsiveness in vitro and in vivo. To define the intracellular pathways involved, we analyzed interleukin (IL)-2R signaling in primary human T cells and in Kit-225 cells. Unlike IL-2-dependent activation of JAK/STAT pathways, activation of Akt kinase was impaired after MV contact both in vitro and in vivo. MV interference with Akt activation was important for immunosuppression, as expression of a catalytically active Akt prevented negative signaling by the MV glycoproteins. Thus, we show here that MV exploits a novel strategy to interfere with T-cell activation during immunosuppression.


Assuntos
Tolerância Imunológica , Vírus do Sarampo/imunologia , Sarampo/imunologia , Proteínas do Leite , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Interleucina-2/metabolismo , Transdução de Sinais/fisiologia , Linfócitos T/imunologia , Androstadienos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Linhagem Celular , Cromonas/farmacologia , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Hemaglutininas Virais/metabolismo , Humanos , Interleucina-2/metabolismo , Janus Quinase 1 , Janus Quinase 3 , Ativação Linfocitária , Sarampo/virologia , Vírus do Sarampo/metabolismo , Vírus do Sarampo/efeitos da radiação , Camundongos , Camundongos Transgênicos , Morfolinas/farmacologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/imunologia , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Sigmodontinae , Baço/citologia , Linfócitos T/enzimologia , Linfócitos T/metabolismo , Transativadores/metabolismo , Proteínas Virais de Fusão/metabolismo , Wortmanina , Proteína de Morte Celular Associada a bcl
7.
J Gen Virol ; 81(Pt 5): 1321-5, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10769075

RESUMO

Plasmids that expressed the nucleocapsid, haemagglutinin and fusion proteins of measles virus (MV) were used to immunize cotton rats (Sigmodon hispidus) against intranasal MV infection. After immunization with all three plasmids, T cell responses and MV-specific antibodies were induced. A reduction in virus titre was observed in lung tissue from animals immunized with plasmids expressing the viral glycoproteins. Histologically, however, a moderate peribronchitis was observed after immunization with the plasmid expressing the fusion protein whereas, after immunization with plasmids expressing haemagglutinin or both glycoproteins, only mild or focal peribronchitis was seen. Immunization with the nucleocapsid did not reduce virus titres, probably because of the failure to induce neutralizing antibodies. A disadvantage of plasmid immunization was its inefficacy in the presence of MV-specific 'maternal' antibodies. This indicates that genetic immunization has to be improved to be a useful alternative vaccine against measles.


Assuntos
Vírus do Sarampo/imunologia , Sarampo/prevenção & controle , Vacinas de DNA , Animais , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática , Hemaglutininas Virais/genética , Hemaglutininas Virais/imunologia , Imunização , Vírus do Sarampo/genética , Testes de Neutralização , Nucleocapsídeo/genética , Nucleocapsídeo/imunologia , Plasmídeos/genética , Sigmodontinae , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
8.
J Gen Virol ; 79 ( Pt 11): 2583-91, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9820133

RESUMO

H-2d mice are resistant to measles virus-induced encephalitis (MVE) and develop Ld-restricted CD8+ T cells which lyse target cells infected with measles virus or with a vaccinia virus recombinant expressing the nucleocapsid protein of measles virus (vvN). In contrast, H-2k mice are susceptible to MVE and generate CD8+ T cells which lyse target cells infected with vvN, but not those infected with MV. We were able to demonstrate that this difference is not due to a defect in the antigen processing machinery, but that Kk molecules require 100-fold more peptide to sensitize target cells for lysis by CTL. vvN replicates well in target cells and therefore enhances the level of epitope peptide available for CTL recognition. In contrast, MV infection is abortive in mouse cells and low levels of epitope peptide are produced. As Ld requires 100-fold less peptide than Kk to sensitize target cells for lysis, the low level of epitope peptide is enough to induce lysis by CD8+ T cells, whereas for recognition via Kk, increased synthesis of protein is required. We propose that the differences in peptide binding between the two H-2 molecules will have consequences for the kinetics of the generation of CD8+ T cells as well as the absolute numbers of CD8+ T cells generated.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Antígenos H-2/imunologia , Vírus do Sarampo/imunologia , Sarampo/imunologia , Animais , Apresentação de Antígeno , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Camundongos
9.
J Virol ; 72(10): 8124-32, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9733853

RESUMO

Nonstructural proteins encoded by measles virus (MV) include the V protein which is translated from an edited P mRNA. V protein is not associated with intracellular or released viral particles and has recently been found to be dispensable for MV propagation in cell culture (H. Schneider, K. Kaelin, and M. A. Billeter, Virology 227:314-322, 1997). Using recombinant MVs (strain Edmonston [ED]) genetically engineered to overexpress V protein (ED-V+) or to be deficient for V protein (ED-V-), we found that in the absence of V both MV-specific proteins and RNAs accumulated to levels higher than those in the parental MV molecular clone (ED-tag), whereas MV-specific gene expression was strongly attenuated in human U-87 glioblastomas cells after infection with ED-V+. The titers of virus released from these cells 48 h after infection with either V mutant virus were lower than those from cells infected with ED-tag. Similarly, significantly reduced titers of infectious virus were reisolated from lung tissue of cotton rats (Sigmodon hispidus) after intranasal infection with both editing mutants compared to titers isolated from ED-tag-infected animals. In cell culture, expression of V protein led to a redistribution of MV N protein in doubly transfected Cos-7 cells, indicating that these proteins form heterologous complexes. This interaction was further confirmed by using a two-hybrid approach with both proteins expressed as Gal4 or VP16 fusion products. Moreover, V protein efficiently competed complexes formed between MV N and P proteins. These findings indicate that V protein acts to balance accumulation of viral gene products in cell culture, and this may be dependent on its interaction with MV N protein. Furthermore, expression of V protein may contribute to viral pathogenicity in vivo.


Assuntos
Vírus do Sarampo/patogenicidade , Fosfoproteínas/genética , RNA Viral/biossíntese , Proteínas Virais/genética , Animais , Humanos , Vírus do Sarampo/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , Ratos , Ratos Endogâmicos , Recombinação Genética , Células Tumorais Cultivadas
10.
J Gen Virol ; 79 ( Pt 5): 1027-31, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9603317

RESUMO

The genes encoding the measles virus (MV) haemagglutinin (H) and fusion (F) proteins were placed under the control of the human cytomegalovirus immediate early promoter in a replication-deficient adenovirus vector. Immunofluorescence and radioimmune precipitation demonstrated the synthesis of each protein and biological activity was confirmed by the detection of haemadsorption and fusion activities in infected cells. Oral as well as parenteral administration of the H-expressing recombinant adenovirus elicited a significant protective response in mice challenged with MV. While the F-expressing adenovirus failed to protect mice, cotton rats immunized with either the H- or F-expressing recombinant showed reduced MV replication in the lungs. Antibodies elicited in mice following immunization with either recombinant had no in vitro neutralizing activity, suggesting a protective mechanism involving a cell-mediated immune response. This study demonstrates the feasibility of using oral administration of adenovirus recombinants to induce protective responses to heterologous proteins.


Assuntos
Adenoviridae , Vírus Defeituosos , Hemaglutininas Virais/imunologia , Vacina contra Sarampo/imunologia , Sarampo/prevenção & controle , Proteínas Virais de Fusão/imunologia , Adenoviridae/fisiologia , Administração Oral , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Linhagem Celular Transformada , Vírus Defeituosos/fisiologia , Feminino , Vetores Genéticos , Hemaglutininas Virais/genética , Humanos , Injeções Intraperitoneais , Vírus do Sarampo/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Ratos , Sigmodontinae , Proteínas Virais de Fusão/genética , Replicação Viral
11.
Virology ; 237(2): 397-403, 1997 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-9356350

RESUMO

Naturally occurring mutations in Human T-cell Leukemia Virus Type 1 (HTLV-1) Tax protein lead to loss of recognition by cytotoxic T-lymphocytes. Most of these mutations also abolish or severely impair the transactivation function of Tax. Ninety percent of the rex gene, which encodes the viral regulator of mRNA splicing (Rex), overlaps with the tax gene. In this paper, we report that four previously described point mutations in tax that abolished CTL recognition and activity did not alter either the dimerisation function or the ability to export viral mRNA of the corresponding Rex proteins. Rex proteins containing two other amino acid changes were likewise functional. However, five Rex deletion mutants, predominantly but not exclusively found in HAM/TSP patients, had all lost these functions. We conclude that, although the Tax protein is subject to strong CTL-mediated selection, there are stronger functional constraints on amino acid variation in Rex. This may limit the variation in the tax/rex nucleotide sequence which results in immune evasion.


Assuntos
Genes pX , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Linfócitos T/imunologia , Células Cultivadas , Citotoxicidade Imunológica/genética , Deleção de Genes , Humanos , Splicing de RNA , RNA Viral/genética , Linfócitos T/virologia
12.
J Virol ; 71(10): 7214-9, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9311794

RESUMO

Immune suppression during measles accounts for most of the morbidity and mortality associated with the virus infection. Experimental study of this phenomenon has been hampered by the lack of a suitable animal model. We have used the cotton rat to demonstrate that mitogen-induced proliferation of spleen cells from measles virus-infected animals is impaired. Proliferation inhibition is seen in all lymphocyte subsets and is not dependent on viral replication. Cells which express the viral glycoproteins (hemagglutinin and fusion protein) transiently by transfection induce proliferation inhibition after intraperitoneal inoculation, whereas application of a recombinant measles virus in which measles virus glycoproteins are replaced with the vesicular stomatitis virus G protein does not have an antiproliferative effect. Therefore, in vivo expression of measles virus glycoproteins is sufficient and necessary to induce inhibition of lymphocyte proliferation.


Assuntos
Linfócitos/imunologia , Vírus do Sarampo/imunologia , Sarampo/imunologia , Proteínas do Envelope Viral/biossíntese , Replicação Viral , Animais , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Tolerância Imunológica , Cinética , Ativação Linfocitária , Linfócitos/virologia , Masculino , Vírus do Sarampo/fisiologia , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , Proteínas Recombinantes/biossíntese , Sigmodontinae , Baço/imunologia , Baço/virologia , Transcrição Gênica , Transfecção
13.
J Gen Virol ; 78 ( Pt 8): 1929-33, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9266990

RESUMO

A mouse model was developed to study the virus-host interaction of molecularly cloned human foamy virus (HFV) in vivo. The infectious process was analysed in two mouse strains, CBA/Ca and C57BL/6J, over a period of 24 weeks by PCR on DNAs from various animal tissues; virus serology was examined by immunoblotting. The infection persisted in both mouse strains and did not induce clinical symptoms. Upon infection of adult CBA/Ca mice HFV became detectable by PCR in an increasing number of organs over time. In contrast, in C57BL/6J mice, after an initial phase of dissemination, viral DNA sequences were found only in a few organs. Interestingly, the different course of infection was accompanied by differences in the antiviral immune response. In particular, C57BL/6J mice were high responders with respect to antibodies to the viral Bet protein, while CBA/Ca mice were low responders.


Assuntos
Primatas/virologia , Infecções por Retroviridae/fisiopatologia , Spumavirus/fisiologia , Replicação Viral , Animais , Animais Recém-Nascidos , Primers do DNA , Feminino , Genes pol , Humanos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Especificidade de Órgãos , Reação em Cadeia da Polimerase , Ratos , Ratos Endogâmicos Lew , Ratos Endogâmicos , Ratos Endogâmicos WKY , Infecções por Retroviridae/imunologia , Especificidade da Espécie , Spumavirus/isolamento & purificação
14.
J Mol Evol ; 42(4): 452-8, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8642614

RESUMO

HTLV-I causes T-cell leukemia and tropical spastic paraparesis (TSP) in a minority of infected people, whereas the majority remain healthy. The virus differs little in sequence between isolates but has been shown to have a quasispecies structure. Using the Nei and Gojobori algorithm, we have shown that the proportion of nonsynonymous to synonymous changes in HTLV-I proviral tax gene sequences from healthy seropositive subjects (Dn/Ds = 0.9 to 1.3) is significantly higher than those from TSP patients (Dn/Ds = 0.3 to 0.6). Here we show that the distinction between healthy seropositives and TSP patients can only be seen with proviral tax sequences, but not with cDNA, the amino-terminal or carboxy-terminal half of tax, or the rex gene. The Dn/Ds ratio of proviral tax sequences was used to analyze two TSP patients with atypical features and to investigate the influence of cytotoxic T cells (CTL) on the viral quasispecies.


Assuntos
Evolução Biológica , Portador Sadio/virologia , Produtos do Gene tax/genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Paraparesia Espástica Tropical/virologia , Portador Sadio/imunologia , Doença Crônica , DNA Complementar , Produtos do Gene rex/genética , Variação Genética , Genoma Viral , Vírus Linfotrópico T Tipo 1 Humano/classificação , Humanos , Paraparesia Espástica Tropical/imunologia , Paraparesia Espástica Tropical/mortalidade , Seleção Genética , Linfócitos T Citotóxicos , Fatores de Tempo
15.
J Virol ; 69(4): 2649-53, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7533860

RESUMO

There is a high degree of intraisolate sequence heterogeneity in the tax gene of human T-cell leukemia virus type I (HTLV-I), although the sequence variation between patients is small compared with that of human immunodeficiency virus type 1. In the present study, we investigated whether naturally occurring amino acid substitutions changed the properties of the Tax protein in two respects: first, recognition of the protein by cytotoxic T lymphocytes (CTL), and second, the ability of the Tax protein to transactivate various promoters. We found that (i) all of the observed amino acid substitutions that occur in known CTL epitopes abolished the recognition of the synthetic peptide representing the respective epitope; (ii) these substitutions occurred significantly more frequently in subjects carrying HLA-A2; and (iii) most of the amino acid substitutions severely reduced the ability of Tax protein to transactivate three promoters: the HTLV-I long terminal repeat, the c-fos promoter, and the interleukin-2 receptor alpha chain promoter.


Assuntos
Produtos do Gene tax/metabolismo , Variação Genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Linfócitos T Citotóxicos/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Epitopos/genética , Produtos do Gene tax/genética , Genes fos , Antígeno HLA-A2/genética , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas
16.
J Virol ; 68(10): 6778-81, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8084014

RESUMO

Human T-cell leukemia virus type I (HTLV-1) causes T-cell leukemia and tropical spastic paraparesis (TSP) in a minority of infected people, whereas the majority remain healthy. No association between a particular HTLV-I sequence and disease manifestation has been found in previous studies. We studied here the sequence variability of the gene for the HTLV-I Tax protein, which is the dominant target antigen of the very strong cytotoxic T-lymphocyte response to the virus. In HTLV-I infection, the intraisolate nucleotide variability is much greater than the variability between isolates. The predicted protein sequence of Tax was significantly more variable in the healthy seropositive individuals' provirus than in those of the patients with TSP. Thus, tax sequence heterogeneity, rather than the presence of particular sequences, distinguishes healthy HTLV-I-seropositive individuals from patients with TSP.


Assuntos
Portador Sadio/microbiologia , Produtos do Gene tax/genética , Genes pX , Variação Genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Paraparesia Espástica Tropical/microbiologia , Sequência de Bases , Primers do DNA , DNA Viral/análise , Vírus Linfotrópico T Tipo 1 Humano/isolamento & purificação , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Homologia de Sequência do Ácido Nucleico
18.
J Virol ; 67(1): 75-81, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8093223

RESUMO

In measles virus (MV) infection in humans, meningitis and encephalitis are important complications. However, little is known of the pathogenesis of MV encephalitis, in particular about the role of the immune response. We have examined the role of cytotoxic T lymphocytes (CTL) in a mouse model of MV-induced encephalitis. We report here that the resistance of inbred strains of mice to MV-induced encephalitis correlated with the major histocompatibility complex (MHC) haplotype and that only resistant mouse strains mounted an effective CTL response to MV. Mice with low susceptibility to MV infection, such as the BALB/c strain (H-2d), generated CTL, whereas the highly susceptible strains, C3H (H-2k) and C57BL/6 (H-2b), revealed very poor CTL responses. MV-induced CTL were usually CD8+, and the generation of these cells was independent of the route of inoculation or the time postinfection. CD4+ T cells were generally only weakly lytic. The nucleocapsid protein was the major target antigen for CTL in BALB/c mice, although in some experiments the hemagglutinin was also recognized. CTL from C3H and C57BL/6 mice did not lyse MV-infected target cells. However, targets infected with vaccinia virus recombinants expressing the nucleocapsid protein or hemagglutinin were lysed, but levels of cytotoxicity were still low. Experiments using target cells transfected with single MHC class I genes suggested inefficient antigen presentation of MV proteins by the MHC molecules of the H-2k and H-2b haplotypes.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Encefalite/imunologia , Sarampo/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Antígenos CD8/imunologia , Capsídeo/imunologia , Citotoxicidade Imunológica , Suscetibilidade a Doenças , Encefalite/etiologia , Antígenos H-2 , Injeções Intraventriculares , Complexo Principal de Histocompatibilidade/imunologia , Sarampo/complicações , Vírus do Sarampo/patogenicidade , Camundongos , Camundongos Endogâmicos , Especificidade da Espécie , Virulência
19.
J Virol ; 65(4): 1695-700, 1991 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1825854

RESUMO

Lewis rats immunized with recombinant vaccinia virus expressing the nucleocapsid (N) protein of measles virus were protected from encephalitis when subsequently challenged by intracerebral infection with neurotropic measles virus. Immunized rats revealed polyvalent antibodies to the N protein of measles virus in the absence of any neutralizing antibodies as well as an N protein-specific proliferative lymphocyte response. Depletion of CD8+ T lymphocytes did not abrogate the protective potential of the N protein-specific cell-mediated immune response in rats, while protection could be adoptively transferred with N protein-specific CD4+ T lymphocytes. These results indicate that a CD4+ cell-mediated immune response specific for the N protein of measles virus is sufficient to control measles virus infections of the central nervous system.


Assuntos
Anticorpos Antivirais/biossíntese , Capsídeo/imunologia , Encefalite/prevenção & controle , Vacina contra Sarampo , Vírus do Sarampo/imunologia , Sarampo/prevenção & controle , Proteínas do Core Viral/imunologia , Animais , Encefalite/complicações , Encefalite/microbiologia , Sarampo/complicações , Ratos , Ratos Endogâmicos Lew , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia , Vaccinia virus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA