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











Base de dados
Intervalo de ano de publicação
1.
Nat Immunol ; 22(3): 370-380, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33574619

RESUMO

During chronic infection and cancer, a self-renewing CD8+ T cell subset maintains long-term immunity and is critical to the effectiveness of immunotherapy. These stem-like CD8+ T cells diverge from other CD8+ subsets early after chronic viral infection. However, pathways guarding stem-like CD8+ T cells against terminal exhaustion remain unclear. Here, we show that the gene encoding transcriptional repressor BACH2 is transcriptionally and epigenetically active in stem-like CD8+ T cells but not terminally exhausted cells early after infection. BACH2 overexpression enforced stem-like cell fate, whereas BACH2 deficiency impaired stem-like CD8+ T cell differentiation. Single-cell transcriptomic and epigenomic approaches revealed that BACH2 established the transcriptional and epigenetic programs of stem-like CD8+ T cells. In addition, BACH2 suppressed the molecular program driving terminal exhaustion through transcriptional repression and epigenetic silencing. Thus, our study reveals a new pathway that enforces commitment to stem-like CD8+ lineage and prevents an alternative terminally exhausted cell fate.


Assuntos
Infecções por Arenaviridae/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Epigênese Genética , Células Precursoras de Linfócitos T/metabolismo , Transcrição Gênica , Animais , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/virologia , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Fatores de Transcrição de Zíper de Leucina Básica/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Linhagem da Célula , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Células Precursoras de Linfócitos T/imunologia , Células Precursoras de Linfócitos T/virologia , Transdução de Sinais
2.
PLoS Pathog ; 16(10): e1008948, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33045019

RESUMO

Pathogenicity often differs dramatically among even closely related arenavirus species. For instance, Junín virus (JUNV), the causative agent of Argentine hemorrhagic fever (AHF), is closely related to Tacaribe virus (TCRV), which is normally avirulent in humans. While little is known about how host cell pathways are regulated in response to arenavirus infection, or how this contributes to virulence, these two viruses have been found to differ markedly in their ability to induce apoptosis. However, details of the mechanism(s) governing the apoptotic response to arenavirus infections are unknown. Here we confirm that TCRV-induced apoptosis is mitochondria-regulated, with associated canonical hallmarks of the intrinsic apoptotic pathway, and go on to identify the pro- and anti-apoptotic Bcl-2 factors responsible for regulating this process. In particular, levels of the pro-apoptotic BH3-only proteins Noxa and Puma, as well as their canonical transcription factor p53, were strongly increased. Interestingly, TCRV infection also led to the accumulation of the inactive phosphorylated form of another pro-apoptotic BH3-only protein, Bad (i.e. as phospho-Bad). Knockout of Noxa or Puma suppressed apoptosis in response to TCRV infection, whereas silencing of Bad increased apoptosis, confirming that these factors are key regulators of apoptosis induction in response to TCRV infection. Further, we found that while the highly pathogenic JUNV does not induce caspase activation, it still activated upstream pro-apoptotic factors, consistent with current models suggesting that JUNV evades apoptosis by interfering with caspase activation through a nucleoprotein-mediated decoy function. This new mechanistic insight into the role that individual BH3-only proteins and their regulation play in controlling apoptotic fate in arenavirus-infected cells provides an important experimental framework for future studies aimed at dissecting differences in the apoptotic responses between arenaviruses, their connection to other cell signaling events and ultimately the relationship of these processes to pathogenesis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Infecções por Arenaviridae/patologia , Arenavirus do Novo Mundo/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Replicação Viral , Proteína de Morte Celular Associada a bcl/metabolismo , Proteínas Reguladoras de Apoptose/genética , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/metabolismo , Infecções por Arenaviridae/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Domínios Proteicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteína de Morte Celular Associada a bcl/genética
3.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31462569

RESUMO

Several mammarenaviruses can cause deadly hemorrhagic fever infections in humans, with limited preventative and therapeutic measures available. Arenavirus cell entry is mediated by the viral glycoprotein (GP) complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The GP2 cytoplasmic tail (CT) is relatively conserved among arenaviruses and is known to interact with the SSP to regulate GP processing and membrane fusion, but its biological role in the context of an infectious virus has not been fully characterized. Using a Pichinde virus (PICV) GP expression vector and a PICV reverse genetics system, we systematically characterized the functional roles of 12 conserved residues within the GP2 CT in GP processing, trafficking, assembly, and fusion, as well as in viral replication. Except for P478A and K505A R508A, alanine substitutions at conserved residues abolished GP processing and membrane fusion in plasmid-transfected cells. Six invariant H and C residues and W503 are essential for viral replication, as evidenced by the fact that their mutant viruses could not be rescued. Both P480A and R482A mutant viruses were rescued, grew similarly to wild-type (WT) virus, and produced evidently processed GP1 and GP2 subunits in virus-infected cells, despite the fact that the same mutations abolished GP processing and membrane fusion in a plasmid-based protein expression system, illustrating the importance of using an infectious-virus system for analyzing viral glycoprotein function. In summary, our results demonstrate an essential biological role of the GP2 CT in arenavirus replication and suggest it as a potential novel target for developing antivirals and/or attenuated viral vaccine candidates.IMPORTANCE Several arenaviruses, such as Lassa virus (LASV), can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, for which no FDA-approved vaccines or therapeutics are available. Viral entry is mediated by the arenavirus GP complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The cytoplasmic tail (CT) of GP2 is highly conserved among arenaviruses, but its functional role in viral replication is not completely understood. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we show that the GP2 CT contains certain conserved residues that are essential for virus replication, implicating it as a potentially good target for developing antivirals and live-attenuated viral vaccines against deadly arenavirus pathogens.


Assuntos
Glicoproteínas/metabolismo , Vírus Pichinde/genética , Proteínas do Envelope Viral/genética , Células A549 , Substituição de Aminoácidos/genética , Animais , Arenaviridae , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/metabolismo , Arenavirus/genética , Arenavirus/metabolismo , Linhagem Celular , Chlorocebus aethiops , Glicoproteínas/genética , Células HEK293 , Humanos , Fusão de Membrana/genética , Mutação/genética , Vírus Pichinde/metabolismo , Sinais Direcionadores de Proteínas/genética , Células Vero , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Replicação Viral
4.
J Virol ; 87(24): 13930-5, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109228

RESUMO

Ocozocoautla de Espinosa virus (OCEV) is a novel, uncultured arenavirus. We found that the OCEV glycoprotein mediates entry into grivet and bat cells through transferrin receptor 1 (TfR1) binding but that OCEV glycoprotein precursor (GPC)-pseudotyped retroviruses poorly entered 53 human cancer cell lines. Interestingly, OCEV and Tacaribe virus could use bat, but not human, TfR1. Replacing three human TfR1 amino acids with their bat ortholog counterparts transformed human TfR1 into an efficient OCEV and Tacaribe virus receptor.


Assuntos
Infecções por Arenaviridae/metabolismo , Infecções por Arenaviridae/veterinária , Arenavirus do Novo Mundo/fisiologia , Quirópteros/metabolismo , Chlorocebus aethiops/metabolismo , Receptores da Transferrina/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/virologia , Arenavirus do Novo Mundo/genética , Linhagem Celular , Quirópteros/genética , Quirópteros/virologia , Chlorocebus aethiops/genética , Chlorocebus aethiops/virologia , Humanos , Dados de Sequência Molecular , Receptores da Transferrina/genética , Receptores Virais/genética , Alinhamento de Sequência , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
5.
Blood ; 121(22): 4473-83, 2013 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-23596046

RESUMO

The precise microRNAs and their target cellular processes involved in generation of durable T-cell immunity remain undefined. Here we show a dynamic regulation of microRNAs as CD8 T cells differentiate from naïve to effector and memory states, with short-lived effectors transiently expressing higher levels of oncogenic miR-17-92 compared with the relatively less proliferating memory-fated effectors. Conditional CD8 T-cell-intrinsic gain or loss of expression of miR-17-92 in mature cells after activation resulted in striking reciprocal effects compared with wild-type counterparts in the same infection milieu-miR-17-92 deletion resulted in lesser proliferation of antigen-specific cells during primary expansion while favoring enhanced IL-7Rα and Bcl-2 expression and multicytokine polyfunctionality; in contrast, constitutive expression of miR-17-92 promoted terminal effector differentiation, with decreased formation of polyfunctional lymphoid memory cells. Increased proliferation upon miR-17-92 overexpression correlated with decreased expression of tumor suppressor PTEN and increased PI3K-AKT-mTOR signaling. Thus, these studies identify miR17-92 as a critical regulator of CD8 T-cell expansion and effector and memory lineages in the physiological context of acute infection, and present miR-17-92 as a potential target for modulating immunologic outcome after vaccination or immunotherapeutic treatments of cancer, chronic infections, or autoimmune disorders.


Assuntos
Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/genética , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica , MicroRNAs/imunologia , Doença Aguda , Animais , Infecções por Arenaviridae/genética , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Proliferação de Células , Feminino , Coriomeningite Linfocítica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Serina-Treonina Quinases TOR/metabolismo , Transcrição Gênica/imunologia , Regulação para Cima/imunologia
6.
J Virol ; 87(11): 6406-14, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536681

RESUMO

The arenaviruses are an important family of emerging viruses that includes several causative agents of severe hemorrhagic fevers in humans that represent serious public health problems. A crucial step of the arenavirus life cycle is maturation of the envelope glycoprotein precursor (GPC) by the cellular subtilisin kexin isozyme 1 (SKI-1)/site 1 protease (S1P). Comparison of the currently known sequences of arenavirus GPCs revealed the presence of a highly conserved aromatic residue at position P7 relative to the SKI-1/S1P cleavage side in Old World and clade C New World arenaviruses but not in New World viruses of clades A and B or cellular substrates of SKI-1/S1P. Using a combination of molecular modeling and structure-function analysis, we found that residue Y285 of SKI-1/S1P, distal from the catalytic triad, is implicated in the molecular recognition of the aromatic "signature residue" at P7 in the GPC of Old World Lassa virus. Using a quantitative biochemical approach, we show that Y285 of SKI-1/S1P is crucial for the efficient processing of peptides derived from Old World and clade C New World arenavirus GPCs but not of those from clade A and B New World arenavirus GPCs. The data suggest that during coevolution with their mammalian hosts, GPCs of Old World and clade C New World viruses expanded the molecular contacts with SKI-1/S1P beyond the classical four-amino-acid recognition sequences and currently occupy an extended binding pocket.


Assuntos
Infecções por Arenaviridae/enzimologia , Arenavirus do Novo Mundo/metabolismo , Arenavirus do Velho Mundo/metabolismo , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/virologia , Arenavirus do Novo Mundo/classificação , Arenavirus do Novo Mundo/genética , Arenavirus do Velho Mundo/classificação , Arenavirus do Velho Mundo/genética , Células CHO , Cricetinae , Humanos , Dados de Sequência Molecular , Pró-Proteína Convertases/química , Pró-Proteína Convertases/genética , Processamento de Proteína Pós-Traducional , Alinhamento de Sequência , Serina Endopeptidases/química , Serina Endopeptidases/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 29(2): 113-7, 2013 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-23388326

RESUMO

Objective To investigate the efficiencies of transfection and expression of human recombinant adenovirus Ad5F35-IL-12 in the different kinds of human mononuclear macrophages. Methods The human recombinant adenovirus Ad5F35-IL-12 was used to infect human peripheral blood monocytes, pleural fluid macrophages as well as THP-1, U937 monocyte cell lines and their phorbol myristate acetate (PMA)-induced macrophages. 48 h later, green fluorescence was observed under the fluorescence microscope to detect the transfection efficiency. The expressions of IL-12 double-subunits (p35, p40) mRNA were tested by RT-PCR and the level of IL-12p70 protein in the cell culture supernatant was detected with ELISA. Results The human recombinant adenovirus Ad5F35-IL-12 successfully infected the human peripheral blood monocytes, pleural fluid macrophages, THP-1 monocytes, U937 monocytes, and THP-1 and U937 macrophages induced with PMA. All above infected mononuclear macrophages effectively secreted IL-12p70 protein, and they were listed from high to low of IL-12p70 protein level as pleural fluid macrophages, U937 and THP-1 macrophages induced with PMA, U937 monocytes, human peripheral blood monocytes and THP-1 monocytes. Conclusion The human recombinant adenovirus Ad5F35-IL-12 could infect different kinds of mononuclear macrophages, and IL-12 p70 protein could be successfully expressed in cell supernatants.


Assuntos
Adenoviridae/genética , Infecções por Arenaviridae/genética , Macrófagos/virologia , Ensaio de Imunoadsorção Enzimática , Humanos , Interleucina-12/biossíntese , Interleucina-12/imunologia , Macrófagos/imunologia , Proteínas Recombinantes/genética
8.
Cell Mol Life Sci ; 70(16): 2849-57, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23115008

RESUMO

Glycosylation of proteins is arguably the most prevalent co- and post-translational modification. It is responsible for increased heterogeneity and functional diversity of proteins. Here we discuss the importance of one type of glycosylation, specifically O-mannosylation and its relationship to a number of human diseases. The most widely studied O-mannose modified protein is alpha-dystroglycan (α-DG). Recent studies have focused intensely on α-DG due to the severity of diseases associated with its improper glycosylation. O-mannosylation of α-DG is involved in cancer metastasis, arenavirus entry, and multiple forms of congenital muscular dystrophy [1, 2]. In this review, we discuss the structural and functional characteristics of O-mannose-initiated glycan structures on α-DG, enzymes involved in the O-mannosylation pathway, and the diseases that are a direct result of disruptions within this pathway.


Assuntos
Infecções por Arenaviridae/metabolismo , Distroglicanas/metabolismo , Manose/metabolismo , Distrofias Musculares/metabolismo , Neoplasias/metabolismo , Animais , Infecções por Arenaviridae/genética , Distroglicanas/química , Distroglicanas/genética , Glicosilação , Humanos , Manose/química , Manose/genética , Distrofias Musculares/genética , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia
9.
Immunity ; 37(1): 158-70, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22705104

RESUMO

Signals from commensal bacteria can influence immune cell development and susceptibility to infectious or inflammatory diseases. However, the mechanisms by which commensal bacteria regulate protective immunity after exposure to systemic pathogens remain poorly understood. Here, we demonstrate that antibiotic-treated (ABX) mice exhibit impaired innate and adaptive antiviral immune responses and substantially delayed viral clearance after exposure to systemic LCMV or mucosal influenza virus. Furthermore, ABX mice exhibited severe bronchiole epithelial degeneration and increased host mortality after influenza virus infection. Genome-wide transcriptional profiling of macrophages isolated from ABX mice revealed decreased expression of genes associated with antiviral immunity. Moreover, macrophages from ABX mice exhibited defective responses to type I and type II IFNs and impaired capacity to limit viral replication. Collectively, these data indicate that commensal-derived signals provide tonic immune stimulation that establishes the activation threshold of the innate immune system required for optimal antiviral immunity.


Assuntos
Bactérias/imunologia , Imunidade Inata , Vírus/imunologia , Imunidade Adaptativa , Animais , Antibacterianos/farmacologia , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/imunologia , Bactérias/efeitos dos fármacos , Suscetibilidade a Doenças/imunologia , Interferons/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia
10.
J Biol Chem ; 286(8): 6192-200, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21159779

RESUMO

Arenaviruses are responsible for acute hemorrhagic fevers worldwide and are recognized to pose significant threats to public health and biodefense. Small molecule compounds have recently been discovered that inhibit arenavirus entry and protect against lethal infection in animal models. These chemically distinct inhibitors act on the tripartite envelope glycoprotein (GPC) through its unusual stable signal peptide subunit to stabilize the complex against pH-induced activation of membrane fusion in the endosome. Here, we report the production and characterization of the intact transmembrane GPC complex of Junín arenavirus and its interaction with these inhibitors. The solubilized GPC is antigenically indistinguishable from the native protein and forms a homogeneous trimer in solution. When reconstituted into a lipid bilayer, the purified complex interacts specifically with its cell-surface receptor transferrin receptor-1. We show that small molecule entry inhibitors specific to New World or Old World arenaviruses bind to the membrane-associated GPC complex in accordance with their respective species selectivities and with dissociation constants comparable with concentrations that inhibit GPC-mediated membrane fusion. Furthermore, competitive binding studies reveal that these chemically distinct inhibitors share a common binding pocket on GPC. In conjunction with previous genetic studies, these findings identify the pH-sensing interface of GPC as a highly vulnerable target for antiviral intervention. This work expands our mechanistic understanding of arenavirus entry and provides a foundation to guide the development of small molecule compounds for the treatment of arenavirus hemorrhagic fevers.


Assuntos
Antígenos CD/metabolismo , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Vírus Junin/metabolismo , Receptores da Transferrina/metabolismo , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/metabolismo , Animais , Antígenos CD/genética , Antivirais/química , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/metabolismo , Chlorocebus aethiops , Humanos , Vírus Junin/genética , Receptores da Transferrina/genética , Células Vero , Proteínas do Envelope Viral/genética , Internalização do Vírus/efeitos dos fármacos
11.
J Autoimmun ; 35(4): 404-13, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20932718

RESUMO

It is not fully understood how the expression level of autoantigens in beta cells impacts autoimmune diabetes (T1D) development. Earlier studies using ovalbumin and also insulin had shown that secreted antigens could enhance diabetes development through facilitated presentation by antigen presenting cells. Here we sought to determine how the expression level of a membrane bound, non-secreted or cross-presented neo-antigen, the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV), would influence T1D. We found that an RIP-LCMV transgenic mouse line exhibiting higher levels of beta cell GP expression developed more severe diabetes after LCMV infection or transfer of high numbers of activated autoreactive T cells. Importantly, all beta cells were lost and a significant increase in morbidity and mortality from T1D was noted. Insulitis and accumulation of autoaggressive CD8 cells was more profound in the RIP-LCMV-GP high-expressor line. Interestingly, the additional introduction of neo-antigen-specific CD4(+) helper or regulatory T cells was able to influence diabetogenesis positively or negatively. We conclude that a higher degree of autoantigen expression results in increased diabetes susceptibility. Therefore, autoantigens such as insulin that are expressed at higher levels in beta cells might have a more profound impact on diabetes pathogenesis.


Assuntos
Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Células Secretoras de Insulina/metabolismo , Vírus da Coriomeningite Linfocítica/fisiologia , Transferência Adotiva , Animais , Antígenos Virais/biossíntese , Antígenos Virais/genética , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/fisiopatologia , Autoantígenos/biossíntese , Autoantígenos/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/transplante , Células Cultivadas , Apresentação Cruzada/genética , Citotoxicidade Imunológica/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatologia , Progressão da Doença , Glicoproteínas/biossíntese , Glicoproteínas/genética , Células Secretoras de Insulina/patologia , Ativação Linfocitária/genética , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética
12.
Cell Physiol Biochem ; 26(3): 263-72, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20798510

RESUMO

More than 500 million people worldwide are persistently infected with either hepatitis B virus (HBV) or hepatitis C virus (HCV). Although both viruses are poorly cytopathic, persistent infection causes severe immunopathologic damage to liver tissue; histologically, such damage is characterized by fatty liver disease, liver fibrosis, and a higher likelihood of hepatocellular carcinoma. Virus-specific CD8+ T cells play a crucial role during infection with hepatitis viruses. On the one hand, rapid activation of CD8+ T cells can control the virus and therefore inhibit its persistence. On the other hand, once the virus persists in the liver, the chronic activation of virus-specific T cells leads to continued liver cell damage. This double-edged role of CD8+ T cells determines the final outcome of infection. In half of cases of human HCV infection, the virus persists; in the other half, the virus is controlled. Additional insights into the molecular mechanisms that determine the course of the disease may be gained from the study of appropriate murine models. This review discusses the similarities and differences between infection with lymphocytic choriomeningitis virus (LCMV) in mice and chronic infection with hepatitis virus in humans.


Assuntos
Infecções por Arenaviridae/genética , Hepatite Viral Animal/genética , Vírus da Coriomeningite Linfocítica , Animais , Infecções por Arenaviridae/imunologia , Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Hepacivirus/imunologia , Vírus da Hepatite B/imunologia , Hepatite Crônica/genética , Hepatite Crônica/imunologia , Hepatite Viral Animal/imunologia , Humanos , Imunidade Inata/genética , Fígado/virologia , Camundongos
13.
J Virol ; 84(19): 9947-56, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20668086

RESUMO

Arenaviruses cause severe human disease ranging from aseptic meningitis following lymphocytic choriomeningitis virus (LCMV) infection to hemorrhagic fever syndromes following infection with Guanarito virus (GTOV), Junin virus (JUNV), Lassa virus (LASV), Machupo virus (MACV), Sabia virus (SABV), or Whitewater Arroyo virus (WWAV). Cellular immunity, chiefly the CD8(+) T-cell response, plays a critical role in providing protective immunity following infection with the Old World arenaviruses LASV and LCMV. In the current study, we evaluated whether HLA class I-restricted epitopes that are cross-reactive among pathogenic arenaviruses could be identified for the purpose of developing an epitope-based vaccination approach that would cross-protect against multiple arenaviruses. We were able to identify a panel of HLA-A*0201-restricted peptides derived from the same region of the glycoprotein precursor (GPC) of LASV (GPC spanning residues 441 to 449 [GPC(441-449)]), LCMV (GPC(447-455)), JUNV (GPC(429-437)), MACV (GPC(444-452)), GTOV (GPC(427-435)), and WWAV (GPC(428-436)) that displayed high-affinity binding to HLA-A*0201 and were recognized by CD8(+) T cells in a cross-reactive manner following LCMV infection or peptide immunization of HLA-A*0201 transgenic mice. Immunization of HLA-A*0201 mice with the Old World peptide LASV GPC(441-449) or LCMV GPC(447-455) induced high-avidity CD8(+) T-cell responses that were able to kill syngeneic target cells pulsed with either LASV GPC(441-449) or LCMV GPC(447-455) in vivo and provided significant protection against viral challenge with LCMV. Through this study, we have demonstrated that HLA class I-restricted, cross-reactive epitopes exist among diverse arenaviruses and that individual epitopes can be utilized as effective vaccine determinants for multiple pathogenic arenaviruses.


Assuntos
Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/prevenção & controle , Arenavirus do Velho Mundo , Vacinas Virais/administração & dosagem , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/virologia , Antígenos Virais/genética , Infecções por Arenaviridae/genética , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , Arenavirus do Velho Mundo/genética , Arenavirus do Velho Mundo/imunologia , Arenavirus do Velho Mundo/patogenicidade , Linfócitos T CD8-Positivos/imunologia , Reações Cruzadas , Citotoxicidade Imunológica , Epitopos/administração & dosagem , Epitopos/genética , Antígenos HLA-A/genética , Antígeno HLA-A2 , Humanos , Vírus Lassa/genética , Vírus Lassa/imunologia , Vírus Lassa/patogenicidade , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Transgênicos , Vacinas Virais/genética , Vacinas Virais/imunologia
14.
J Immunol ; 183(10): 6554-60, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19846867

RESUMO

TNF has been shown to be important for controlling many pathogens. Here, we directly demonstrate using wild-type TNF(-/-) and TNFR1(-/-) mice that TNF does play a role in protection against vaccinia virus (VV) infection in naive mice. Since VV replication is also partially controlled in lymphocytic choriomeningitis virus (LCMV)-immune C57BL/6J mice through the process of heterologous immunity, we questioned whether TNF was required in mediating this protection. VV-infected LCMV-immune mice that were TNF-deficient as a consequence of genetic deletion or receptor blockade demonstrated normal recruitment and selective expansion of cross-reactive LCMV-specific memory CD8 T cells and controlled VV infection similar to LCMV-immune mice having TNF function. This indicates that neither TNF nor lymphotoxin, which uses the same receptor, was required in mediating protective heterologous immunity against VV. Indeed, prior immunity to LCMV made the role of TNF in protection against VV infection much less important, even under conditions of lethal dose inoculum. Thus, heterologous immunity may help explain why treatment of patients with anti-TNF compounds is reasonably well tolerated with relatively few infectious complications.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/imunologia , Vaccinia virus/imunologia , Vacínia/imunologia , Animais , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/virologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Memória Imunológica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Linfotoxina-alfa/imunologia , Linfotoxina-alfa/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Vacínia/genética , Vacínia/virologia
15.
J Immunol ; 181(11): 7786-99, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19017968

RESUMO

The CD3gamma di-leucine-based motif plays a central role in TCR down-regulation. However, little is understood about the role of the CD3gamma di-leucine-based motif in physiological T cell responses. In this study, we show that the expansion in numbers of virus-specific CD8(+) T cells is impaired in mice with a mutated CD3gamma di-leucine-based motif. The CD3gamma mutation did not impair early TCR signaling, nor did it compromise recruitment or proliferation of virus-specific T cells, but it increased the apoptosis rate of the activated T cells by increasing down-regulation of the antiapoptotic molecule Bcl-2. This resulted in a 2-fold reduction in the clonal expansion of virus-specific CD8(+) T cells during the acute phase of vesicular stomatitis virus and lymphocytic choriomeningitis virus infections. These results identify an important role of CD3gamma-mediated TCR down-regulation in virus-specific CD8(+) T cell responses.


Assuntos
Infecções por Arenaviridae/imunologia , Complexo CD3/imunologia , Linfócitos T CD8-Positivos/imunologia , Regulação para Baixo/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Infecções por Rhabdoviridae/imunologia , Vesiculovirus/imunologia , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/imunologia , Animais , Infecções por Arenaviridae/genética , Complexo CD3/genética , Camundongos , Camundongos Transgênicos , Mutação/imunologia , Peptídeos/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Receptores de Antígenos de Linfócitos T/genética , Infecções por Rhabdoviridae/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Proteínas Virais/imunologia
16.
J Immunol ; 176(11): 6665-72, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16709825

RESUMO

Immunoproteasome subunits low-molecular mass polypeptide (LMP)2 and LMP7 affect Ag presentation by MHC class I molecules. In the present study, we investigated the function of the third immunosubunit LMP10/multicatalytic endopeptidase complex-like (MECL)-1 (beta2i) in MECL-1 gene-targeted mice. The number of CD8+ splenocytes in MECL-1-/- mice was 20% lower than in wild-type mice. Infection with lymphocytic choriomeningitis virus (LCMV) elicited a markedly reduced cytotoxic T cell (CTL) response to the LCMV epitopes GP276-286/Db and NP205-212/Kb in MECL-1-/- mice. The weak CTL response to GP276-286/Db was not due to an impaired generation of this epitope but was attributed to a decreased precursor frequency of GP276-286/Db-specific T cells. The expansion of TCR-Vbeta10+ T cells, which contain GP276-286/Db-specific cells, was reduced in LCMV-infected MECL-1-/- mice. Taken together, our data reveal an in vivo function of MECL-1 in codetermining the T cell repertoire for an antiviral CTL response.


Assuntos
Cisteína Endopeptidases/deficiência , Cisteína Endopeptidases/genética , Subpopulações de Linfócitos T/enzimologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/genética , Infecções por Arenaviridae/enzimologia , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Caspases/metabolismo , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/fisiologia , Citotoxicidade Imunológica/genética , Marcação de Genes , Humanos , Epitopos Imunodominantes/imunologia , Fígado/enzimologia , Fígado/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Linfopenia/enzimologia , Linfopenia/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Subpopulações de Linfócitos T/transplante , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia
17.
J Immunol ; 170(5): 2382-9, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12594261

RESUMO

Targeted disruption of T cell costimulatory pathways, particularly CD28 and CD40, has allowed for the development of minimally myeloablative strategies for the induction of mixed allogeneic chimerism and donor-specific tolerance across full MHC barriers. In this study we analyze in depth the ability of mixed allogeneic chimeras in two strain combinations to mount effective host-restricted and donor-restricted antiviral CD4 and CD8 responses, as well as the impact of development of mixed chimerism on the maintenance of pre-existing memory populations. While antiviral CD8 responses in mixed chimeras following acute viral infection with lymphocytic choriomeningitis virus Armstrong or vaccinia virus are largely host-restricted, donor-restricted CD8 responses as well as host- and donor-restricted CD4 responses are also readily detected, and virus is promptly cleared. We further demonstrate that selection of donor-restricted T cells in mixed chimeras is principally mediated by bone marrow-derived cells in the thymus. Conversely, we find that mixed chimeras exhibit a deficit in their ability to deal with a chronic lymphocytic choriomeningitis virus clone 13 infection. Encouragingly, pre-existing memory populations are largely unaffected by the development of high level mixed chimerism and maintain the ability to control viral rechallenge. Our results suggest that while pre-existing T cell memory and primary immunocompetence to acute infection are preserved in mixed allogeneic chimeras, MHC class I and/or class II tissue matching may be required to fully preserve immunocompetence in dealing with chronic viral infections.


Assuntos
Imunocompetência/genética , Isoantígenos/genética , Quimera por Radiação/imunologia , Animais , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/virologia , Transplante de Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Hematopoese/genética , Hematopoese/imunologia , Humanos , Imunidade Inata/genética , Imunização Secundária , Imunocompetência/imunologia , Memória Imunológica/genética , Isoantígenos/imunologia , Ativação Linfocitária/genética , Vírus da Coriomeningite Linfocítica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Células Tumorais Cultivadas , Vacínia/genética , Vacínia/imunologia , Vacínia/virologia , Vaccinia virus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA