Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Viruses ; 15(1)2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36680046

RESUMO

The Department of Defense recently began an effort to improve and standardize virus challenge materials and efficacy determination strategies for testing therapeutics and vaccines. This includes stabilization of virus genome sequences in cDNA form where appropriate, use of human-derived virus isolates, and noninvasive strategies for determination of challenge virus replication. Eventually, it is desired that these approaches will satisfy the FDA "Animal Rule" for licensure, which substitutes animal efficacy data when human data are unlikely to be available. To this end, we created and examined the virulence phenotype of cDNA clones of prototypic human infection-derived strains of the alphaviruses, Venezuelan (VEEV INH9813), eastern (EEEV V105) and western (WEEV Fleming) equine encephalitis viruses, and created fluorescent and luminescent reporter expression vectors for evaluation of replication characteristics in vitro and in vivo. Sequences of minimally passaged isolates of each virus were used to synthesize full-length cDNA clones along with a T7 transcription promoter-based bacterial propagation vector. Viruses generated from the cDNA clones were compared with other "wild type" strains derived from cDNA clones and GenBank sequences to identify and eliminate putative tissue culture artifacts accumulated in the cell passaged biological stocks. This was followed by examination of aerosol and subcutaneous infection and disease in mouse models. A mutation that increased heparan sulfate binding was identified in the VEEV INH9813 biological isolate sequence and eliminated from the cDNA clone. Viruses derived from the new human isolate cDNA clones showed similar mouse virulence to existing clone-derived viruses after aerosol or subcutaneous inoculation.


Assuntos
Vírus da Encefalite Equina Venezuelana , Vírus da Encefalite Equina do Oeste , Estados Unidos , Humanos , Animais , Cavalos , Camundongos , DNA Complementar/genética , Fenótipo , Células Clonais
2.
PLoS Pathog ; 15(10): e1007867, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31658290

RESUMO

Eastern equine encephalitis virus (EEEV), a mosquito-borne RNA virus, is one of the most acutely virulent viruses endemic to the Americas, causing between 30% and 70% mortality in symptomatic human cases. A major factor in the virulence of EEEV is the presence of four binding sites for the hematopoietic cell-specific microRNA, miR-142-3p, in the 3' untranslated region (3' UTR) of the virus. Three of the sites are "canonical" with all 7 seed sequence residues complimentary to miR-142-3p while one is "non-canonical" and has a seed sequence mismatch. Interaction of the EEEV genome with miR-142-3p limits virus replication in myeloid cells and suppresses the systemic innate immune response, greatly exacerbating EEEV neurovirulence. The presence of the miRNA binding sequences is also required for efficient EEEV replication in mosquitoes and, therefore, essential for transmission of the virus. In the current studies, we have examined the role of each binding site by point mutagenesis of the seed sequences in all combinations of sites followed by infection of mammalian myeloid cells, mosquito cells and mice. The resulting data indicate that both canonical and non-canonical sites contribute to cell infection and animal virulence, however, surprisingly, all sites are rapidly deleted from EEEV genomes shortly after infection of myeloid cells or mice. Finally, we show that the virulence of a related encephalitis virus, western equine encephalitis virus, is also dependent upon miR-142-3p binding sites.


Assuntos
Regiões 3' não Traduzidas/genética , Vírus da Encefalite Equina do Leste/genética , Vírus da Encefalite Equina do Oeste/genética , MicroRNAs/genética , Replicação Viral/genética , Aedes , Animais , Sítios de Ligação/genética , Linhagem Celular , Cricetinae , Vírus da Encefalite Equina do Leste/imunologia , Vírus da Encefalite Equina do Leste/patogenicidade , Vírus da Encefalite Equina do Oeste/imunologia , Vírus da Encefalite Equina do Oeste/patogenicidade , Encefalomielite Equina/imunologia , Encefalomielite Equina/virologia , Feminino , Imunidade Inata/imunologia , Células L , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Virulência/genética
3.
PLoS Pathog ; 15(2): e1007584, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30742691

RESUMO

Live attenuated vaccines (LAVs), if sufficiently safe, provide the most potent and durable anti-pathogen responses in vaccinees with single immunizations commonly yielding lifelong immunity. Historically, viral LAVs were derived by blind passage of virulent strains in cultured cells resulting in adaptation to culture and a loss of fitness and disease-causing potential in vivo. Mutations associated with these phenomena have been identified but rarely have specific attenuation mechanisms been ascribed, thereby limiting understanding of the attenuating characteristics of the LAV strain and applicability of the attenuation mechanism to other vaccines. Furthermore, the attenuated phenotype is often associated with single nucleotide changes in the viral genome, which can easily revert to the virulent sequence during replication in animals. Here, we have used a rational approach to attenuation of eastern equine encephalitis virus (EEEV), a mosquito-transmitted alphavirus that is among the most acutely human-virulent viruses endemic to North America and has potential for use as an aerosolized bioweapon. Currently, there is no licensed antiviral therapy or vaccine for this virus. Four virulence loci in the EEEV genome were identified and were mutated individually and in combination to abrogate virulence and to resist reversion. The resultant viruses were tested for virulence in mice to examine the degree of attenuation and efficacy was tested by subcutaneous or aerosol challenge with wild type EEEV. Importantly, all viruses containing three or more mutations were avirulent after intracerebral infection of mice, indicating a very high degree of attenuation. All vaccines protected from subcutaneous EEEV challenge while a single vaccine with three mutations provided reproducible, near-complete protection against aerosol challenge. These results suggest that informed mutation of virulence determinants is a productive strategy for production of LAVs even with highly virulent viruses such as EEEV. Furthermore, these results can be directly applied to mutation of analogous virulence loci to create LAVs from other viruses.


Assuntos
Vírus da Encefalite Equina do Leste/genética , Vírus da Encefalite Equina do Leste/imunologia , Vacinas Atenuadas/biossíntese , Animais , Anticorpos Neutralizantes , Linhagem Celular , Cricetinae , Vírus da Encefalite Equina do Leste/patogenicidade , Encefalomielite Equina do Leste/veterinária , Encefalomielite Equina do Leste/virologia , Feminino , Engenharia Genética/métodos , Cavalos , Camundongos , Mutação , América do Norte , Projetos de Pesquisa , Vacinas Atenuadas/imunologia , Vacinas Virais/biossíntese , Virulência , Fatores de Virulência
4.
Nat Microbiol ; 4(1): 187-197, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30455470

RESUMO

Eastern equine encephalitis virus (EEEV) is a mosquito-transmitted alphavirus with a high case mortality rate in humans. EEEV is a biodefence concern because of its potential for aerosol spread and the lack of existing countermeasures. Here, we identify a panel of 18 neutralizing murine monoclonal antibodies (mAbs) against the EEEV E2 glycoprotein, several of which have 'elite' activity with 50 and 99% effective inhibitory concentrations (EC50 and EC99) of less than 10 and 100 ng ml-1, respectively. Alanine-scanning mutagenesis and neutralization escape mapping analysis revealed epitopes for these mAbs in domains A or B of the E2 glycoprotein. A majority of the neutralizing mAbs blocked infection at a post-attachment stage, with several inhibiting viral membrane fusion. Administration of one dose of anti-EEEV mAb protected mice from lethal subcutaneous or aerosol challenge. These experiments define the mechanistic basis for neutralization by protective anti-EEEV mAbs and suggest a path forward for treatment and vaccine design.


Assuntos
Anticorpos Monoclonais/imunologia , Vírus da Encefalite Equina do Leste/imunologia , Encefalomielite Equina/imunologia , Encefalomielite Equina/prevenção & controle , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Chlorocebus aethiops , Cricetinae , Encefalomielite Equina/virologia , Mapeamento de Epitopos , Epitopos/imunologia , Feminino , Células HEK293 , Humanos , Camundongos , Domínios Proteicos/imunologia , Células Vero
5.
mSphere ; 3(5)2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30232164

RESUMO

Type I interferon (IFN)-stimulated genes (ISGs) have critical roles in inhibiting virus replication and dissemination. Despite advances in understanding the molecular basis of ISG restriction, the antiviral mechanisms of many remain unclear. The 20-kDa ISG ISG20 is a nuclear 3'-5' exonuclease with preference for single-stranded RNA (ssRNA) and has been implicated in the IFN-mediated restriction of several RNA viruses. Although the exonuclease activity of ISG20 has been shown to degrade viral RNA in vitro, evidence has yet to be presented that virus inhibition in cells requires this activity. Here, we utilized a combination of an inducible, ectopic expression system and newly generated Isg20-/- mice to investigate mechanisms and consequences of ISG20-mediated restriction. Ectopically expressed ISG20 localized primarily to Cajal bodies in the nucleus and restricted replication of chikungunya and Venezuelan equine encephalitis viruses. Although restriction by ISG20 was associated with inhibition of translation of infecting genomic RNA, degradation of viral RNAs was not observed. Instead, translation inhibition of viral RNA was associated with ISG20-induced upregulation of over 100 other genes, many of which encode known antiviral effectors. ISG20 modulated the production of IFIT1, an ISG that suppresses translation of alphavirus RNAs. Consistent with this observation, the pathogenicity of IFIT1-sensitive alphaviruses was increased in Isg20-/- mice compared to that of wild-type viruses but not in cells ectopically expressing ISG20. Our findings establish an indirect role for ISG20 in the early restriction of RNA virus replication by regulating expression of other ISGs that inhibit translation and possibly other activities in the replication cycle.IMPORTANCE The host immune responses to infection lead to the production of type I interferon (IFN), and the upregulation of interferon-stimulated genes (ISGs) reduces virus replication and virus dissemination within a host. Ectopic expression of the interferon-induced 20-kDa exonuclease ISG20 suppressed replication of chikungunya virus and Venezuelan equine encephalitis virus, two mosquito-vectored RNA alphaviruses. Since the replication of alphavirus genomes occurs exclusively in the cytoplasm, the mechanism of nucleus-localized ISG20 inhibition of replication is unclear. In this study, we determined that ISG20 acts as a master regulator of over 100 genes, many of which are ISGs. Specifically, ISG20 upregulated IFIT1 genes and inhibited translation of the alphavirus genome. Furthermore, IFIT1-sensitive alphavirus replication was increased in Isg20-/- mice compared to the replication of wild-type viruses but not in cells ectopically expressing ISG20. We propose that ISG20 acts as an indirect regulator of RNA virus replication in the cytoplasm through the upregulation of many other ISGs.


Assuntos
Exonucleases/genética , Exorribonucleases/genética , Interferon Tipo I/genética , Replicação Viral , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Linhagem Celular , Vírus Chikungunya/fisiologia , Vírus da Encefalite Equina Venezuelana/fisiologia , Feminino , Interações Hospedeiro-Patógeno , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , RNA Viral/análise , Proteínas de Ligação a RNA , Regulação para Cima
6.
Curr Opin Virol ; 23: 30-34, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28288385

RESUMO

Alphaviruses are medically important mosquito-borne viruses that cause a range of diseases in humans from febrile illness to arthritis or encephalitis. The innate immune response functions to suppress virus replication through upregulation of antiviral molecules and contributes to development of the adaptive immune response. Myeloid cells act as master regulators of virus infection by initiating both the innate and adaptive immune responses. Alphaviruses are capable of antagonizing individual components of these responses to increase replicative fitness in vivo. However, recently, studies have demonstrated that some alphaviruses avoid myeloid cell replication altogether to achieve a similar effect. In this review, we summarize how alphaviruses evade myeloid cell infection and individual inductive mechanisms, thereby limiting the activation of the innate immune response.


Assuntos
Alphavirus/imunologia , Alphavirus/patogenicidade , Proliferação de Células , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Células Mieloides/fisiologia , Células Mieloides/virologia , Imunidade Adaptativa , Animais , Humanos , Imunidade Inata , Células Mieloides/imunologia
7.
Trends Mol Med ; 23(1): 80-93, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27989642

RESUMO

microRNAs (miRNAs) are non-coding RNAs that regulate many processes within a cell by manipulating protein levels through direct binding to mRNA and influencing translation efficiency, or mRNA abundance. Recent evidence demonstrates that miRNAs can also affect RNA virus replication and pathogenesis through direct binding to the RNA virus genome or through virus-mediated changes in the host transcriptome. Here, we review the current knowledge on the interaction between RNA viruses and cellular miRNAs. We also discuss how cell and tissue-specific expression of miRNAs can directly affect viral pathogenesis. Understanding the role of cellular miRNAs during viral infection may lead to the identification of novel mechanisms to block RNA virus replication or cell-specific regulation of viral vector targeting.


Assuntos
MicroRNAs/genética , Infecções por Vírus de RNA/genética , Vírus de RNA/fisiologia , RNA Viral/genética , Replicação Viral , Animais , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , MicroRNAs/metabolismo , Infecções por Vírus de RNA/metabolismo , Infecções por Vírus de RNA/patologia , Infecções por Vírus de RNA/virologia , Vírus de RNA/genética , RNA Viral/metabolismo , Transcriptoma
8.
Methods Mol Biol ; 1428: 127-37, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27236796

RESUMO

The ability to transfect synthetic mRNAs into cells to measure processes such as translation efficiency or mRNA decay has been an invaluable tool in cell biology. The use of electroporation over other methods of transfection is an easy, inexpensive, highly efficient, and scalable method to introduce synthetic mRNA into a wide range of cell types. More recently, coupling of noncoding RNA sequences or protein coding regions from viral pathogens to fluorescent or bioluminescence proteins in RNA "reporters" has permitted study of host-pathogen interactions. These can range from virus infection of cells to translation of the viral genome, replication and stability of viral RNAs, or the efficacy of host antiviral responses. In this chapter, we describe a method for electroporating viral RNA reporters into both fibroblastic and myeloid cells that encode firefly or Renilla luciferase, whose reaction with specific substrates and light emitting activity is a measure of viral RNA translation efficiency. We have used this method to examine host interferon-dependent responses that inhibit viral translation along with identifying secondary structures in the 5' nontranslated region (NTR) and microRNA binding sites in the 3' NTR that are responsible for antagonizing the host innate immune responses and restricting viral cell tropism.


Assuntos
Alphavirus/imunologia , Eletroporação/métodos , Fibroblastos/imunologia , Células Mieloides/imunologia , RNA Viral/genética , Alphavirus/genética , Animais , Linhagem Celular , Cricetinae , Imunidade Inata , Interferons/metabolismo , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Luciferases de Renilla/genética , Luciferases de Renilla/metabolismo , RNA Viral/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
9.
Virus Res ; 206: 99-107, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-25630058

RESUMO

The non-coding regions found at the 5' and 3' ends of alphavirus genomes regulate viral gene expression, replication, translation and virus-host interactions, which have significant implications for viral evolution, host range, and pathogenesis. The functions of these non-coding regions are mediated by a combination of linear sequence and structural elements. The capped 5' untranslated region (UTR) contains promoter elements, translational regulatory sequences that modulate dependence on cellular translation factors, and structures that help to avoid innate immune defenses. The polyadenylated 3' UTR contains highly conserved sequence elements for viral replication, binding sites for cellular miRNAs that determine cell tropism, host range, and pathogenesis, and conserved binding regions for a cellular protein that influences viral RNA stability. Nonetheless, there are additional conserved elements in non-coding regions of the virus (e.g., the repeated sequence elements in the 3' UTR) whose function remains obscure. Thus, key questions remain as to the function of these short yet influential untranslated segments of alphavirus RNAs.


Assuntos
Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Alphavirus/fisiologia , Regulação Viral da Expressão Gênica , RNA Viral/genética , RNA Viral/metabolismo , Replicação Viral , Alphavirus/genética , Interações Hospedeiro-Patógeno , Ligação Proteica , Biossíntese de Proteínas , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Proteínas Virais/metabolismo
10.
Am J Obstet Gynecol ; 212(1): 71.e1-71.e8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25108145

RESUMO

OBJECTIVE: Primary human trophoblasts were previously shown to be resistant to viral infection, and able to confer this resistance to nontrophoblast cells. Can trophoblasts protect nontrophoblastic cells from infection by viruses or other intracellular pathogens that are implicated in perinatal infection? STUDY DESIGN: Isolated primary term human trophoblasts were cultured for 48-72 hours. Diverse nonplacental human cell lines (U2OS, human foreskin fibroblast, TZM-bl, MeWo, and Caco-2) were preexposed to either trophoblast conditioned medium, nonconditioned medium, or miR-517-3p for 24 hours. Cells were infected with several viral and nonviral pathogens known to be associated with perinatal infections. Cellular infection was defined and quantified by plaque assays, luciferase assays, microscopy, and/or colonization assays. Differences in infection were assessed by Student t test or analysis of variance with Bonferroni correction. RESULTS: Infection by rubella and other togaviruses, human immunodeficiency virus-1, and varicella zoster was attenuated in cells preexposed to trophoblast-conditioned medium (P < .05), and a partial effect by the chromosome 19 microRNA miR-517-3p on specific pathogens. The conditioned medium had no effect on infection by Toxoplasma gondii or Listeria monocytogenes. CONCLUSION: Our findings indicate that medium conditioned by primary human trophoblasts attenuates viral infection in nontrophoblastic cells. Our data point to a trophoblast-specific antiviral effect that may be exploited therapeutically.


Assuntos
Resistência à Doença , Doenças Fetais/virologia , Trofoblastos/fisiologia , Viroses/imunologia , Células Cultivadas , Meios de Cultivo Condicionados , Humanos , Recém-Nascido
11.
Expert Rev Vaccines ; 13(12): 1423-5, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25073901

RESUMO

Eastern equine encephalitis virus (EEEV) is a highly neurovirulent mosquito-borne alphavirus that causes severe morbidity and mortality upon human infection. Recent emergence of EEEV into nonendemic regions in the USA and Panama demonstrates the need for the development of an effective EEEV vaccine for licensure for human use. The current EEEV vaccine is available to only at-risk laboratory workers but is poorly immunogenic and requires multiple boosters. In this editorial, we summarize recent developments in understanding alphavirus virulence mechanisms that could be utilized to rationally design a live attenuated vaccine against EEEV or other alphaviruses.


Assuntos
Infecções por Alphavirus/veterinária , Infecções por Alphavirus/virologia , Vírus de RNA/imunologia , Vírus de RNA/fisiologia , Vacinas Virais/imunologia , Vacinas Virais/isolamento & purificação , Fatores de Virulência/metabolismo , Infecções por Alphavirus/prevenção & controle , Animais , Pesquisa Biomédica/tendências , Descoberta de Drogas/tendências , Humanos , Vírus de RNA/patogenicidade
12.
Science ; 343(6172): 783-7, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24482115

RESUMO

Although interferon (IFN) signaling induces genes that limit viral infection, many pathogenic viruses overcome this host response. As an example, 2'-O methylation of the 5' cap of viral RNA subverts mammalian antiviral responses by evading restriction of Ifit1, an IFN-stimulated gene that regulates protein synthesis. However, alphaviruses replicate efficiently in cells expressing Ifit1 even though their genomic RNA has a 5' cap lacking 2'-O methylation. We show that pathogenic alphaviruses use secondary structural motifs within the 5' untranslated region (UTR) of their RNA to alter Ifit1 binding and function. Mutations within the 5'-UTR affecting RNA structural elements enabled restriction by or antagonism of Ifit1 in vitro and in vivo. These results identify an evasion mechanism by which viruses use RNA structural motifs to avoid immune restriction.


Assuntos
Infecções por Alphavirus/imunologia , Alphavirus/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Capuzes de RNA/química , Capuzes de RNA/imunologia , RNA Viral/química , RNA Viral/imunologia , Regiões 5' não Traduzidas/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Alphavirus/fisiologia , Infecções por Alphavirus/virologia , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Conformação de Ácido Nucleico , Proteínas de Ligação a RNA , Replicação Viral
13.
Nature ; 506(7487): 245-8, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24352241

RESUMO

Currently, there is little evidence for a notable role of the vertebrate microRNA (miRNA) system in the pathogenesis of RNA viruses. This is primarily attributed to the ease with which these viruses mutate to disrupt recognition and growth suppression by host miRNAs. Here we report that the haematopoietic-cell-specific miRNA miR-142-3p potently restricts the replication of the mosquito-borne North American eastern equine encephalitis virus in myeloid-lineage cells by binding to sites in the 3' non-translated region of its RNA genome. However, by limiting myeloid cell tropism and consequent innate immunity induction, this restriction directly promotes neurologic disease manifestations characteristic of eastern equine encephalitis virus infection in humans. Furthermore, the region containing the miR-142-3p binding sites is essential for efficient virus infection of mosquito vectors. We propose that RNA viruses can adapt to use antiviral properties of vertebrate miRNAs to limit replication in particular cell types and that this restriction can lead to exacerbation of disease severity.


Assuntos
Vírus da Encefalite Equina do Leste/imunologia , Vírus da Encefalite Equina do Leste/patogenicidade , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata/imunologia , MicroRNAs/genética , Regiões 3' não Traduzidas/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Sítios de Ligação/genética , Linhagem Celular , Cricetinae , Culicidae/virologia , Modelos Animais de Doenças , Vírus da Encefalite Equina do Leste/genética , Vírus da Encefalite Equina do Leste/crescimento & desenvolvimento , Feminino , Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune/genética , Imunidade Inata/genética , Insetos Vetores/virologia , Masculino , Camundongos , MicroRNAs/metabolismo , Células Mieloides/imunologia , Células Mieloides/virologia , Especificidade de Órgãos , Replicação Viral/genética , Replicação Viral/imunologia
14.
Eur J Immunol ; 40(5): 1315-27, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20213733

RESUMO

Memory cross-reactive CD8+ T-cell responses may induce protection or immunopathology upon secondary viral challenge. To elucidate the potential role of T cells in sequential flavivirus infection, we characterized cross-reactive CD4+ and CD8+ T-cell responses between attenuated and pathogenic Japanese encephalitis virus (JEV) and pathogenic West Nile virus (WNV). A previously reported WNV NS4b CD8+ T-cell epitope and its JEV variant elicited CD8+ T-cell responses in both JEV- and WNV-infected mice. The peptide variant homologous to the immunizing virus induced greater cytokine secretion and activated higher frequencies of epitope-specific CD8+ T cells. However, there was a virus-dependent, peptide variant-independent pattern of cytokine secretion; the IFNgamma+-to-IFNgamma+TNFalpha+ CD8+ T-cell ratio was greater in JEV- than in WNV-infected mice. Despite similarities in viral burden for pathogenic WNV and JEV viruses, CD8+ T cells from pathogenic JEV-immunized mice exhibited functional and phenotypic profiles similar to those seen for the attenuated JEV strain. Patterns of killer cell lectin-like receptor G1 (KLRG1) and CD127 expression differed by virus type, with a rapid expansion and contraction of short-lived effector cells in JEV infection and persistence of high levels of short-lived effector cells in WNV infection. Such cross-reactive T-cell responses to primary infection may affect the outcomes of sequential flavivirus infections.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus da Encefalite Japonesa (Espécie)/imunologia , Infecções por Flavivirus/imunologia , Subpopulações de Linfócitos T/imunologia , Vírus do Nilo Ocidental/imunologia , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Reações Cruzadas , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Epitopos de Linfócito T/imunologia , Feminino , Infecções por Flavivirus/virologia , Imunização , Epitopos Imunodominantes/imunologia , Interferon gama/metabolismo , Linfocinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Oligopeptídeos/imunologia , Organismos Livres de Patógenos Específicos , Subpopulações de Linfócitos T/metabolismo , Vacinas Atenuadas , Carga Viral , Vacinas Virais/imunologia , Replicação Viral , Vírus do Nilo Ocidental/patogenicidade , Vírus do Nilo Ocidental/fisiologia
15.
Biomed Sci Instrum ; 42: 308-13, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16817626

RESUMO

Prior studies show that purified T cell receptors (TCRs) and antibodies catalyze the oxidation of water to H2O2 in the presence of singlet oxygen, but the comparative efficiencies of TCRs and antibodies in this process have not been reported. Since H2O2 has been shown to activate TCRs and selectively regulate redox sensitive TCR signaling pathways, it is important to understand the physiological significance of these recently uncovered processes. This new information might be used to develop new therapeutic tools for immune and inflammatory diseases. In this paper, we present data showing that under equivalent conditions Jurkat T cell membranes produce H2O2 at a rate of 457 pM/min/mg protein/muW/cm2 while IgG antibodies produce H2O2 at a rate of 192 pM/min/mg protein/muW/cm2. Taking into account the number of catalytic sites in a milligram of T cell membranes and IgGs, we calculate that TCRs catalyze H2O2 production at a specific rate that is about 10(6) times greater than the rate of IgGs. Based on these observations and calculations, we conclude that the comparatively high rate of H2O2 production by TCRs makes it more likely that this is a physiologically relevant process than the H2O2 production by IgGs. In addition, the catalytic rate for H2O2 production by TCRs is comparable to the rates of other physiologically important processes, such as catalysis by enzymes. This suggests that singlet oxygen-dependent, TCR mediated, H2O2 production is likely to be physiologically important, perhaps as H2O2 being a small molecule regulator of TCR signal transduction or a modulator of T cell gene transcription.


Assuntos
Anticorpos/metabolismo , Água Corporal/metabolismo , Membrana Celular/metabolismo , Peróxido de Hidrogênio/metabolismo , Modelos Biológicos , Linfócitos T/metabolismo , Água/metabolismo , Anticorpos/imunologia , Membrana Celular/imunologia , Simulação por Computador , Humanos , Peróxido de Hidrogênio/imunologia , Células Jurkat , Oxirredução , Transdução de Sinais/fisiologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA