Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Interferon Cytokine Res ; 42(5): 193-194, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35447043
6.
Viral Immunol ; 30(9): 628-632, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29028182

RESUMO

Viral sequence integration into the mammalian genome has long been perceived as a health risk. In some cases, integration translates to chronic viral infection, and in other instances, oncogenic gene mutations occur. However, research also shows that animal cells can benefit from integrated viral sequences (e.g., to support host cell development or to silence foreign invaders). Here we propose that, comparable with the clustered regularly interspaced short palindromic repeats that provide bacteria with adaptive immunity against invasive bacteriophages, animal cells may co-opt integrated viral sequences to support immune memory. We hypothesize that host cells express viral peptides from open reading frames in integrated sequences to boost adaptive B cell and T cell responses long after replicating viruses are cleared. In support of this hypothesis, we examine previous literature describing (1) viruses that infect acutely (e.g., vaccinia viruses and orthomyxoviruses) followed by unexplained, long-term persistence of viral nucleotide sequences, viral peptides, and virus-specific adaptive immunity, (2) the high frequency of endogenous viral genetic elements found in animal genomes, and (3) mechanisms with which animal host machinery supports foreign sequence integration.


Assuntos
Imunidade Adaptativa/imunologia , DNA Viral/genética , Orthomyxoviridae/genética , Orthomyxoviridae/imunologia , RNA Viral/genética , Vaccinia virus/genética , Vaccinia virus/imunologia , Integração Viral/genética , Imunidade Adaptativa/genética , Animais , Linfócitos B/imunologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Genoma/genética , Humanos , Fases de Leitura Aberta/genética , Orthomyxoviridae/patogenicidade , Linfócitos T/imunologia , Vaccinia virus/patogenicidade
10.
Viral Immunol ; 27(10): 484-96, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25479178

RESUMO

Immunopathologic examination of the lungs of mice with experimental influenza virus infection reveals three prominent findings. (i) There is rapidly developing perivascular (arterial) and peribronchial infiltration with T-cells and invasion of T-cells into the bronchiolar epithelium, separation of epithelial cells from each other and from the basement membrane, leading to defoliation of the bronchial epithelium. This reaction is analogous to a viral exanthema of the skin, such as measles and smallpox. This previously described but unappreciated reaction most likely is an effective way to eliminate virus-infected cells, but may contribute to acute toxicity and mortality. (ii) After this, there is formation of dense collections of lymphocytes adjacent to bronchi consisting mainly of B-cells, with a scattering of T-cells and macrophages. This is known as induced bronchial-associated lymphoid tissue (iBALT) and correlates with increased interleukin (IL)-17 in the lung. iBALT provides sites for a local immune reaction in the lung to both the original infection and related viral infections (heterologous immunity). (iii) Within the first 2-3 weeks, there is proliferation of type II pneumocytes and/or terminal bronchial epithelial cells extending from the terminal bronchioles into the adjacent alveoli, eventually leading to large zones of the lung filled with tumor-like epithelial cells with squamous metaplasia. The proliferation correlates with IL-17 and IL-22 expression, and the extent of this reaction appears to be determined by the availability of T-regulatory cells.


Assuntos
Células Epiteliais Alveolares/fisiologia , Proliferação de Células , Tecido Linfoide/patologia , Infecções por Orthomyxoviridae/patologia , Mucosa Respiratória/patologia , Linfócitos T Citotóxicos/imunologia , Animais , Brônquios/imunologia , Brônquios/patologia , Modelos Animais de Doenças , Pulmão/imunologia , Pulmão/patologia , Tecido Linfoide/imunologia , Camundongos , Infecções por Orthomyxoviridae/imunologia , Mucosa Respiratória/imunologia
12.
J Immunol ; 193(12): 5827-34, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25378595

RESUMO

The oncogenic γ-herpesviruses EBV and Kaposi sarcoma-associated herpesvirus are ubiquitous human pathogens that establish lifelong latent infections maintained by intermittent viral reactivation and reinfection. Effector CD4 T cells are critical for control of viral latency and in immune therapies for virus-associated tumors. In this study, we exploited γHV68 infection of mice to enhance our understanding of the CD4 T cell response during γ-herpesvirus infection. Using a consensus prediction approach, we identified 16 new CD4 epitope-specific responses that arise during lytic infection. An additional epitope encoded by the M2 protein induced uniquely latency-associated CD4 T cells, which were not detected at the peak of lytic infection but only during latency and were not induced postinfection with a latency-deficient virus. M2-specific CD4 T cells were selectively cytotoxic, produced multiple antiviral cytokines, and sustained IL-2 production. Identification of latency-associated cytolytic CD4 T cells will aid in dissecting mechanisms of CD4 immune control of γ-herpesvirus latency and the development of therapeutic approaches to control viral reactivation and pathology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citotoxicidade Imunológica , Epitopos de Linfócito T/imunologia , Gammaherpesvirinae/imunologia , Latência Viral , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/metabolismo , Citocinas/biossíntese , Epitopos de Linfócito T/química , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Interferon gama/biossíntese , Ativação Linfocitária/imunologia , Camundongos , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia
15.
J Virol ; 88(14): 7862-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24789784

RESUMO

CD8 and CD4 T cells are each critically important for immune control of murine gammaherpesvirus 68 (γHV68) infection. In immunocompetent mice, acute γHV68 infection results in lifelong latency, but in the absence of CD4 T cell help, mice succumb to viral recrudescence and disease. However, the requirements for CD4 T cell help in the generation and maintenance of antiviral CD8 T cell responses are incompletely understood, and it is unclear whether there are epitope-specific differences in the requirement of CD8 T cells for CD4 help. In this report, we characterized the CD8 T cell response to γHV68 in major histocompatibility complex (MHC) class II(-/-) mice, which lack CD4 T cells, or after antibody-mediated depletion of CD4 T cells. All antiviral CD8 T cells exhibited marked upregulation of surface expression of the inhibitory receptor programmed death-1 (PD-1), but surprisingly, while the immunodominant memory response appeared to be functionally impaired, helpless CD8 T cells of a subdominant specificity had increased numbers and enhanced functionality. Thus, we demonstrate differential requirements for CD4 help in the antiviral CD8 T cell response to a latent gammaherpesvirus. Importance: γHV68 is a mouse pathogen closely related to the oncogenic human γHVs, which infect a majority of the world's population. Reactivation of these viruses from latency can lead to complications, disease, and even death. CD4 T cells are required for complete immune control of long-term infection, in part by providing key signals to dendritic cells that in turn instruct optimal antiviral CD8 T cell responses. We have investigated multiple virus-specific CD8 T cell responses during infection and identified a subdominant CD8 T cell response that is numerically and functionally enhanced in the absence of CD4 T cell help. This occurs in spite of high surface expression of an inhibitory receptor and in contrast to the immunodominant response, which is impaired. Our data suggest that signals from CD4 T cells are important in maintaining the CD8 T cell hierarchy during γHV infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Herpesviridae/imunologia , Rhadinovirus/imunologia , Animais , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos/química , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Morte Celular Programada 1/análise
17.
J Autoimmun ; 42: 71-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23245703

RESUMO

Human herpesviruses establish lifelong latency. Viral recrudescence can lead to the development of cancers, immunoproliferative disorders, transplantation complications, and thrombocytopenia. Although platelet-specific autoantibodies have been reported in patients infected with the Epstein-Barr virus (EBV), the mechanisms by which thrombocytopenia is induced remain unclear, as do the relative contributions of lytic viral replication and latent viral gene expression. The human gammaherpesviruses are tightly restricted in their ability to infect other mammals, so they are difficult to study in live animal models. Here we show that infection of mice with murine gammaherpesvirus-68 (γHV68), a rodent-specific pathogen closely related to EBV, induces the production of platelet-binding antibodies and causes thrombocytopenia. Infection of antibody-deficient mice does not lead to thrombocytopenia, indicating the platelet decrease is mediated by antibody. Additionally, infection with a latency-null recombinant γHV68 does not induce thrombocytopenia, suggesting factors associated with viral latency drive the infection-induced antibody-mediated thrombocytopenia. These studies describe an important animal model of gammaherpesvirus-induced autoimmune thrombocytopenia and demonstrate that this pathology is mediated by antibody and dependent on viral latency. This model will allow studies of the underlying mechanisms of disease progression and the testing of therapeutic strategies for the alleviation of virus-induced thrombocytopenia.


Assuntos
Anticorpos/imunologia , Transtornos Plaquetários/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 4/imunologia , Latência Viral , Animais , Transtornos Plaquetários/etiologia , Plaquetas/imunologia , Células Cultivadas , Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/complicações , Feminino , Infecções por Herpesviridae/complicações , Humanos , Cadeias mu de Imunoglobulina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade da Espécie , Replicação Viral
18.
J Immunol ; 188(8): 3812-9, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22407914

RESUMO

The γ-herpesviruses are characterized by their ability to establish lifelong latency. Subsequent immune suppression leads to viral reactivation from latency and the onset of a variety of pathologies, including lymphoproliferative disease and cancers. CD8 T cells play a key role in preventing reactivation of latent virus. Therefore, to develop effective therapeutic immune strategies, it is essential to understand the maintenance of CD8 T cell responses during latency. Because the γ-herpesviruses are highly species-specific and mice cannot be infected with the human pathogens, EBV or Kaposi's sarcoma-associated herpesvirus, we have used a natural rodent γ-herpesvirus experimental infection model, γ-herpesvirus-68. In this report, we show that during long-term latent infection, naive CD8 T cells are recruited into the ongoing immune response in an epitope-specific manner. When virus reactivation is induced in vivo, the recruitment of CD8 T cells for some, but not all, epitopes is enhanced. The variation in recruitment is not due to differences in epitope presentation. We also show that CD8 T cells that are newly stimulated during reactivation are functionally impaired compared with acutely stimulated cells in terms of cytokine production. Thus, our results demonstrate unexpected complexity in the response of CD8 T cells specific for different viral epitopes that were stimulated during acute infection, quiescent latency, and reactivation.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Ativação Linfocitária/imunologia , Ativação Viral/imunologia , Latência Viral/imunologia , Transferência Adotiva , Animais , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Citocinas/biossíntese , Citocinas/imunologia , Epitopos de Linfócito B/imunologia , Feminino , Infecções por Herpesviridae/virologia , Especificidade de Hospedeiro , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
Clin Vaccine Immunol ; 19(5): 757-65, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22398245

RESUMO

Vitamin A deficiency (VAD) has profound effects on immune responses in the gut, but its effect on other mucosal responses is less well understood. Sendai virus (SeV) is a candidate human parainfluenza virus type 1 (hPIV-1) vaccine and a candidate vaccine vector for other respiratory viruses. A single intranasal dose of SeV elicits a protective immune response against hPIV-1 within days after vaccination. To define the effect of VAD on acute responses toward SeV, we monitored both antibodies and CD8(+) T cells in mice. On day 10 following SeV infection, there was a trend toward lower antibody activities in the nasal washes of VAD mice than in those of controls, while bronchoalveolar lavage (BAL) fluid and serum antibodies were not reduced. In contrast, there was a dramatic reduction of immunodominant CD8(+) T cell frequencies in the lower respiratory tract (LRT) airways of VAD animals. These T cells also showed unusually high CD103 (the αE subunit of αEß7) expression patterns. In both VAD and control mice, E-cadherin (the ligand for αEß7) was better expressed among epithelial cells lining the upper respiratory tract (URT) than in LRT airways. The results support a working hypothesis that the high CD103 expression among T cell populations in VAD mice alters mechanisms of T cell cross talk with URT and LRT epithelial cells, thereby inhibiting T cell migration and egress into the lower airway. Our data emphasize that the consequences of VAD are not limited to gut-resident cells and characterize VAD influences on an immune response to a respiratory virus vaccine.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/análise , Sistema Respiratório/imunologia , Deficiência de Vitamina A/imunologia , Animais , Anticorpos Antivirais/análise , Sangue/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Linfócitos T CD8-Positivos/química , Endoglina , Células Epiteliais/imunologia , Feminino , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Nasal/química , Mucosa Nasal/imunologia , Gravidez , Vírus Sendai/imunologia
20.
J Immunol ; 188(3): 1049-56, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22198955

RESUMO

The human γ-herpesviruses EBV and Kaposi's sarcoma-associated herpesvirus establish lifelong latent infections, can reactivate in immunocompromised individuals, and are associated with the development of malignancies. Murine γ-herpesvirus-68 (γHV68), a rodent pathogen related to EBV and Kaposi's sarcoma-associated herpesvirus, provides an important model to dissect mechanisms of immune control and investigate vaccine strategies. Infection of mice with γHV68 elicits robust antiviral immunity, and long-term protection from γHV68 reactivation requires both cellular and humoral immune responses. Vaccination of mice with AC-replication and transcription activator (RTA), a highly lytic latency-null recombinant γHV68, results in complete protection from wild-type γHV68 infection that lasts for at least 10 mo. In this report, we examine the immune correlates of AC-RTA-mediated protection and show that sterilizing immunity requires both T cells and Ab. Importantly, Ab was also critical for mitigating viral infection in the brain, and in the absence of Ab-mediated control, amplification of the AC-RTA virus in the brain resulted in fatality. Our results highlight important considerations in the development of vaccination strategies based on live-attenuated viruses.


Assuntos
Anticorpos Antivirais , Rhadinovirus , Vacinas/imunologia , Viroses/imunologia , Latência Viral , Animais , Imunidade , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Vacinação , Vacinas Atenuadas , Ativação Viral , Viroses/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA