Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 10(1): 2121, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31073183

RESUMO

Pulmonary immune control is crucial for protection against pathogens. Here we identify a pathway that promotes host responses during pulmonary bacterial infection; the expression of CD200 receptor (CD200R), which is known to dampen pulmonary immune responses, promotes effective clearance of the lethal intracellular bacterium Francisella tularensis. We show that depletion of CD200R in mice increases in vitro and in vivo infectious burden. In vivo, CD200R deficiency leads to enhanced bacterial burden in neutrophils, suggesting CD200R normally limits the neutrophil niche for infection. Indeed, depletion of this neutrophil niche in CD200R-/- mice restores F. tularensis infection to levels seen in wild-type mice. Mechanistically, CD200R-deficient neutrophils display significantly reduced reactive oxygen species production (ROS), suggesting that CD200R-mediated ROS production in neutrophils is necessary for limiting F. tularensis colonisation and proliferation. Overall, our data show that CD200R promotes the antimicrobial properties of neutrophils and may represent a novel antibacterial therapeutic target.


Assuntos
Francisella tularensis/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Glicoproteínas de Membrana/imunologia , Neutrófilos/imunologia , Tularemia/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Francisella tularensis/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/microbiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Cultura Primária de Células , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Tularemia/microbiologia
2.
Mucosal Immunol ; 10(4): 1021-1030, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27966555

RESUMO

Triggering receptor expressed on myeloid cells-1 (TREM-1) is expressed on neutrophils and monocyte/macrophages and amplifies Toll-like receptor-mediated inflammation during infection. TREM-1 also exists in an antagonistic soluble form (sTREM-1) that has been used as a peripheral biomarker in sepsis, though the mechanisms of its release are not entirely clear. The requirement of TREM-1 in single microbial infections is controversial, with some studies showing a protective role and others a contribution to immunopathology. Furthermore, the role of membrane-bound and sTREM-1 in polygenic infections is currently unknown. In a mouse co-infection model where preceding viral infection greatly enhances bacteria co-infection, we now determine a mechanisms for the striking increase in sTREM-1 and the loss of TREM-1 on surface of neutrophils. We identified a matrix metalloproteinase (MMP)-9 cleavage site in TREM-1 and that the increase of MMP-9 in bronchoalveolar lavage fluid mirrors sTREM-1 release. In vitro studies with neutrophils and MMP-9 and the reduction of sTREM-1 in vivo after MMP-9 inhibition verifies that this enzyme cleaves TREM-1. Intriguingly, MMP-9 inhibition significantly reduces bacterial load and ensuing immunopathology in a co-infection model. This highlights MMP-9 inhibition as a potential therapeutic via blocking cleavage of TREM-1.


Assuntos
Antibacterianos/uso terapêutico , Vírus da Influenza A Subtipo H1N1/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Neutrófilos/imunologia , Infecções por Orthomyxoviridae/metabolismo , Fenilpropionatos/uso terapêutico , Infecções Pneumocócicas/metabolismo , Streptococcus pneumoniae/fisiologia , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo , Administração Intranasal , Animais , Carga Bacteriana/efeitos dos fármacos , Células Cultivadas , Coinfecção , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções Pneumocócicas/tratamento farmacológico , Domínios Proteicos , Proteólise/efeitos dos fármacos
3.
Mucosal Immunol ; 7(3): 489-500, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24064672

RESUMO

Antigen-presenting cell (APC) activation is enhanced by vaccine adjuvants. Most vaccines are based on the assumption that adjuvant activity of Toll-like receptor (TLR) agonists depends on direct, functional activation of APCs. Here, we sought to establish whether TLR stimulation in non-hematopoietic cells contributes to flagellin's mucosal adjuvant activity. Nasal administration of flagellin enhanced T-cell-mediated immunity, and systemic and secretory antibody responses to coadministered antigens in a TLR5-dependent manner. Mucosal adjuvant activity was not affected by either abrogation of TLR5 signaling in hematopoietic cells or the presence of flagellin-specific, circulating neutralizing antibodies. We found that flagellin is rapidly degraded in conducting airways, does not translocate into lung parenchyma and stimulates an early immune response, suggesting that TLR5 signaling is regionalized. The flagellin-specific early response of lung was regulated by radioresistant cells expressing TLR5 (particularly the airway epithelial cells). Flagellin stimulated the epithelial production of a small set of mediators that included the chemokine CCL20, which is known to promote APC recruitment in mucosal tissues. Our data suggest that (i) the adjuvant activity of TLR agonists in mucosal vaccination may require TLR stimulation of structural cells and (ii) harnessing the effect of adjuvants on epithelial cells can improve mucosal vaccines.


Assuntos
Imunidade nas Mucosas , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Receptor 5 Toll-Like/metabolismo , Imunidade Adaptativa , Administração Intranasal , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Linhagem Celular , Flagelina/administração & dosagem , Flagelina/imunologia , Flagelina/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Imunidade nas Mucosas/genética , Imunidade nas Mucosas/imunologia , Camundongos , Camundongos Knockout , Proteólise , Mucosa Respiratória/citologia , Transdução de Sinais , Receptor 5 Toll-Like/genética
4.
Mucosal Immunol ; 5(5): 524-34, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22549744

RESUMO

The lung is colonized by commensal bacteria, some of which are associated with asthma exacerbations. Using the intranasal house-dust mite-sensitized mouse model of allergic airway disease, we show an imbalance in novel antibacterial pathways that culminates in a reduction in neutrophil recruitment to the airspaces and leads to bacterial invasion and dissemination. The expression of TREM (Triggering Receptor Expressed on Myeloid cells)-1 that amplifies Toll-like receptor (TLR) signaling and TREM-2 that inhibits this process is reversed. Furthermore, endogenous TLR inhibitors (A20, Tollip, SOCS1, and IRAK-M) and proteins involved in receptor recycling (TRIAD3) are raised. Consequently, the production of neutrophil chemoattractants is reduced. Intranasal administration of either chemokine restores the ability to recruit neutrophils, which prevents bacterial invasion. A background of allergic airway disease therefore exacerbates bacterial infection by altering key antibacterial innate immune pathways that are amenable to therapeutic intervention.


Assuntos
Pulmão/imunologia , Neutrófilos/imunologia , Infecções Pneumocócicas/imunologia , Hipersensibilidade Respiratória/imunologia , Streptococcus pneumoniae/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Pulmão/microbiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/microbiologia , Infecções Pneumocócicas/complicações , Pyroglyphidae/imunologia , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Hipersensibilidade Respiratória/complicações , Transdução de Sinais , Receptores Toll-Like/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides
5.
Scand J Immunol ; 71(6): 393-402, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20500691

RESUMO

Dendritic cells (DC) are an essential link between the innate and adaptive immune response. To become effective antigen-presenting cells DC need to undergo maturation, during which they up-regulate co-stimulatory molecules and produce cytokines. There is great interest in utilizing DC in vaccination regimes. Over recent years, Toll-like receptor (TLR) signalling has been recognized to be one of the major inducers of DC maturation. This study describes a mutant version of the TLR adaptor molecule MyD88 (termed MyD88lpr) as a novel adjuvant for vaccination regimes. MyD88lpr specifically activates DC by disrupting a DC intrinsic inhibitory mechanism, which is dependent on single immunoglobulin IL-1R-related. Moreover, MyD88lpr was able to induce an IgG2a-dominated response to a co-expressed antigen, suggesting Th1 immunity. However, when used as a vaccine adjuvant for Influenza nucleoprotein there was no significant difference in the lung viral titres during the infection. This study describes MyD88lpr as a potential adjuvant for vaccinations, which would be able to target DC specifically.


Assuntos
Células Dendríticas/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Receptores de Interleucina-1/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antivirais/sangue , Células Dendríticas/efeitos dos fármacos , Feminino , Humanos , Imunidade Inata/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/farmacologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/farmacologia , Organismos Livres de Patógenos Específicos , Células Th1/imunologia , Vacinação/métodos
6.
Mucosal Immunol ; 1(4): 265-78, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19079188

RESUMO

Inflammatory lung disease to innocuous antigens or infectious pathogens is a common occurrence and in some cases, life threatening. Often, the inflammatory infiltrate that accompanies these events contributes to pathology by deleterious effects on otherwise healthy tissue and by compromising lung function by consolidating (blocking) the airspaces. A fine balance, therefore, exists between a lung immune response and immune-mediated damage, and in some the "threshold of ignorance" may be set too low. In most cases, the contributing, potentially offending, cell population or immune pathway is known, as are factors that regulate them. Why then are targeted therapeutic strategies to manipulate them not more commonplace in clinical medicine? This review highlights immune homeostasis in the lung, how and why this is lost during acute lung infection, and strategies showing promise as future immune therapeutics.


Assuntos
Pneumonia/imunologia , Infecções Respiratórias/imunologia , Doença Aguda , Animais , Sobrevivência Celular/fisiologia , Citocinas/imunologia , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Humanos , Imunidade Inata , Tecido Linfoide/imunologia , Macrófagos Alveolares/imunologia , Pneumonia/terapia , Sistema Respiratório/imunologia , Infecções Respiratórias/terapia
7.
Clin Exp Allergy ; 38(6): 1016-24, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18498543

RESUMO

BACKGROUND: The mechanisms underlying exacerbation of asthma induced by respiratory syncytial virus (RSV) infection have been extensively studied in human and animal models. However, most of these studies focused on acute inflammation and little is known of its long-term consequences on remodelling of the airway tissue. OBJECTIVE: The aim of the study was to use a murine model of prolonged allergen-induced airway inflammation to investigate the effect of RSV infection on allergic airway inflammation and tissue remodelling. METHODS: We subjected mice to RSV infection before or during the chronic phase of airway challenges with OVA and compared parameters of airway inflammation and remodelling at the end-point of the prolonged allergen-induced airway inflammation protocol. RESULTS: RSV infection did not affect the severity of airway inflammation in any of the groups studied. However, RSV infection provoked airway remodelling in non-sensitized, allergen-challenged mice that did not otherwise develop any of the features of allergic airways disease. Increased collagen synthesis in the lung and thickening of the bronchial basal membrane was observed in non-sensitized allergen-challenged mice only after prior RSV infection. In addition, fibroblast growth factor (FGF)-2 but not TGF-beta(1) was increased in this group following RSV infection. CONCLUSION: Our data show for the first time that RSV infection can prime the lung of mice that are not previously systemically sensitized, to develop airway remodelling in response to allergen upon sole exposure via the airways. Moreover, our results implicate RSV-induced FGF-2 in the remodelling process in vivo.


Assuntos
Asma/patologia , Pulmão/patologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios , Compostos de Alúmen/administração & dosagem , Análise de Variância , Animais , Asma/imunologia , Asma/virologia , Líquido da Lavagem Broncoalveolar/imunologia , Colágeno/metabolismo , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Imuno-Histoquímica , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/patologia , Estatísticas não Paramétricas , Fator de Crescimento Transformador beta/metabolismo
8.
Parasitology ; 135(7): 825-39, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18477416

RESUMO

Although co-infection is the norm in most human and animal populations, clinicians currently have no practical tool to assist them in choosing the best treatment strategy for such patients. Given the vast range of potential pathogens which may co-infect the host, obtaining such a practical tool may seem an intractable problem. In ecology the joint concepts of functional groups and guilds have been used to conceptually simplify complex ecosystems, in order to understand how their component parts interact and may be manipulated. Here we propose a mechanism by which to apply these concepts to pathogen co-infection systems. Further, we describe how these groups could be incorporated into a mathematical modelling framework which, after validation, could be used as a clinical tool to predict the outcome of any particular combination of pathogens co-infecting a host.


Assuntos
Comorbidade , Ecossistema , Interações Hospedeiro-Parasita/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Modelos Biológicos , Animais , Interações Hospedeiro-Parasita/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Matemática , Dinâmica Populacional , Valor Preditivo dos Testes , Especificidade da Espécie
9.
Biochem Soc Trans ; 34(Pt 6): 1032-6, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17073744

RESUMO

Inflammatory cascades are initiated in response to alarm signals that may result from infection, malignant transformation or trauma. Immunity, however, must be controlled; otherwise damage may occur to otherwise healthy tissue within the same microenvironment. Similarly, peripheral tolerance mechanisms must ensure that autoreactive thymic or bone marrow emigrants do not respond upon encounter with the autoantigen. Organized lymphoid structures such as lymph nodes, spleen and Peyer's patches appear to regulate inflammation successfully, displaying controlled expansion and contraction. However, when immune cells flood into effector sites, the organization of T- and B-lymphocytes is lacking. What controls inflammatory cascades in lymph nodes but rarely in effector sites is not clear. We believe the difference lies in the Toll-like receptor ligand load, which is high in effector sites and drives uncontrolled inflammation. Similarly, we believe that initiation of autoimmune inflammation is initiated by the liberation of inflammatory signals due to infection or trauma. In this review, we highlight some of the molecules responsible for maintaining an activated T-cell phenotype, strategies to interrupt these therapeutically and the impact of ligating inhibitory receptors on antigen-presenting cells.


Assuntos
Inflamação/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos CD/imunologia , Proteínas Fetais/imunologia , Humanos , Imunidade , Memória Imunológica , Influenza Humana/imunologia , Camundongos , Modelos Imunológicos , Proteínas Tirosina Quinases/imunologia
12.
J Immunol Methods ; 257(1-2): 155-61, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11687249

RESUMO

The study of target cell lysis and cytokine production are valuable tools to characterize antigen-specific T and NK cell function during virus infections. After localized infections in compartments such as the lung or the brain, however, cell numbers isolated from these organs are too low to perform standard assays with individual mice. Here, we report a few simple modifications of the classical 51Cr release assay allowing reduction of the number of required effector cells by a factor of 10 without loosing sensitivity or specificity. Using not more than 4x10(5) effector cells, we were able to study ex vivo virus-specific CTL or NK activity from the lungs of individual mice after infection with respiratory syncytial virus (RSV) and from the brains of mice infected with Borna disease virus (BDV). Flow cytometric analysis of interferon-gamma production by virus-specific T cells including appropriate controls was achieved with as few as 10(5) effector cells.


Assuntos
Testes Imunológicos de Citotoxicidade/métodos , Linfócitos T Citotóxicos/imunologia , Animais , Doença de Borna/imunologia , Doença de Borna/patologia , Encéfalo/citologia , Encéfalo/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Linhagem Celular , Citometria de Fluxo , Técnicas In Vitro , Células Matadoras Naturais/imunologia , Pulmão/citologia , Pulmão/imunologia , Contagem de Linfócitos , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia
13.
Eur J Immunol ; 31(9): 2566-73, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11536154

RESUMO

TNF antagonists are effective treatments for rheumatoid arthritis and Crohn's disease, and have been tried with variable success in other diseases caused by immune damage. To test the hypothesis that viral lung diseases caused by respiratory syncytial virus or influenza virus are partly due to overproduction of TNF, we used anti-TNF antibody to treat mice with lung disease caused by these viruses. TNF depletion reduced pulmonary recruitment of inflammatory cells, cytokine production by T cells and the severity of illness without preventing virus clearance. These broad beneficial effects suggest that TNF antagonists might be tested as treatments of human viral lung diseases.


Assuntos
Bronquiolite Viral/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Anticorpos/farmacologia , Anticorpos Antivirais/biossíntese , Bronquiolite Viral/patologia , Bronquiolite Viral/virologia , Citocinas/biossíntese , Feminino , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Eosinofilia Pulmonar/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Redução de Peso
14.
Eur J Immunol ; 31(9): 2574-82, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11536155

RESUMO

In mice acutely infected with respiratory syncytial virus (RSV), more than 20% of pulmonary CD8(+) T cells, but only 2-3% of CD8(+) T cells in the draining lymph node secreted interferon-gamma in response to a single peptide. Surprisingly, the percentage of virus-specific T cells in the lung remained at these high levels long after the acute infection. Pulmonary memory T cells were further studied in a sensitive adoptive transfer system, which allows visualizing polyclonal CD4(+) and CD8(+) virus-specific memory T cell responses. Fifty days after infection, persisting RSV-specific pulmonary T cells remained CD69(hi) CD62L(lo), but had returned to a resting memory state according to functional criteria. In the absence of neutralizing antibodies reinfection first induced cell division among virus-specific memory T cells 3 days after infection predominantly in the local lymph node. However, divided cells then rapidly accumulated in the lung without significantly increasing in the lymph node. These results suggest rapid export of reactivated cells from the lymph node to the target organ. Thus, although memory T cells can be maintained in the infected organ after a localized virus infection, amplification of a recall response appears to be most effective in organized lymphoid tissue.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Pneumopatias/virologia , Ativação Linfocitária , Infecções por Vírus Respiratório Sincicial/imunologia , Transferência Adotiva , Animais , Antígenos Virais/imunologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/transplante , Células Cultivadas , Interferon gama/biossíntese , Cinética , Pulmão/imunologia , Pneumopatias/imunologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus Sinciciais Respiratórios/imunologia
15.
Nat Med ; 7(6): 732-7, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11385512

RESUMO

Elevated expression of heat-shock proteins (HSPs) can benefit a microbial pathogen struggling to penetrate host defenses during infection, but at the same time might provide a crucial signal alerting the host immune system to its presence. To determine which of these effects predominate, we constructed a mutant strain of Mycobacterium tuberculosis that constitutively overexpresses Hsp70 proteins. Although the mutant was fully virulent in the initial stage of infection, it was significantly impaired in its ability to persist during the subsequent chronic phase. Induction of microbial genes encoding HSPs might provide a novel strategy to boost the immune response of individuals with latent tuberculosis infection.


Assuntos
Proteínas de Bactérias , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Mycobacterium tuberculosis/fisiologia , Tuberculose/microbiologia , Animais , Eletroforese em Gel de Poliacrilamida , Expressão Gênica , Proteínas de Choque Térmico/genética , Humanos , Interferon gama/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Mycobacterium bovis/genética , Mycobacterium bovis/fisiologia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Proteínas Repressoras/genética , Baço/imunologia , Baço/metabolismo , Temperatura , Tuberculose/imunologia , Tuberculose/patologia
16.
J Exp Med ; 193(7): 785-92, 2001 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-11283151

RESUMO

T cells secreting interleukin (IL)-4 and IL-5 (T helper cell type 2 [Th2] cells) play a detrimental role in a variety of diseases, but specific methods of regulating their activity remain elusive. T1/ST2 is a surface ligand of the IL-1 receptor family, expressed on Th2- but not on interferon (IFN)-gamma-producing Th1 cells. Prior exposure of BALB/c mice to the attachment (G) or fusion (F) protein of respiratory syncytial virus (RSV) increases illness severity during intranasal RSV challenge, due to Th2-driven lung eosinophilia and exuberant Th1-driven pulmonary infiltration, respectively. We used these polar models of viral illness to study the recruitment of T1/ST2 cells to the lung and to test the effects of anti-T1/ST2 treatment in vivo. T1/ST2 was present on a subset of CD4(+) cells from mice with eosinophilic lung disease. Monoclonal anti-T1/ST2 treatment reduced lung inflammation and the severity of illness in mice with Th2 (but not Th1) immunopathology. These results show that inhibition of T1/ST2 has a specific effect on virally induced Th2 responses and suggests that therapy targeted at this receptor might be of value in treating Th2-driven illness.


Assuntos
Proteínas de Membrana , Proteínas/imunologia , Receptores de Interleucina-1/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Linfócitos T Auxiliares-Indutores , Animais , Eosinofilia/imunologia , Eosinofilia/terapia , Feminino , Proteína 1 Semelhante a Receptor de Interleucina-1 , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Interleucina , Infecções por Vírus Respiratório Sincicial/terapia , Células Th1 , Células Th2
17.
J Immunol ; 166(2): 1106-13, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11145691

RESUMO

In an effort to develop a safe and effective vaccine against respiratory syncytial virus (RSV), we used Escherichia coli heat-labile toxin (LT), and LTK63 (an LT mutant devoid of ADP-ribosyltransferase activity) to elicit murine CD8(+) CTL responses to an intranasally codelivered CTL peptide from the second matrix protein (M2) of RSV. M2(82-90)-specific CD8(+) T cells were detected by IFN-gamma enzyme-linked immunospot and (51)Cr release assay in local and systemic lymph nodes, and their induction was dependent on the use of a mucosal adjuvant. CTL elicited by peptide immunization afforded protection against RSV challenge, but also enhanced weight loss. CTL-mediated viral clearance was not dependent on IFN-gamma since depletion using specific mAb during RSV challenge did not affect cellular recruitment or viral clearance. Depletion of IFN-gamma did, however, reduce the concentration of TNF detected in lung homogenates of challenged mice and largely prevented the weight loss associated with CTL-mediated viral clearance. Mice primed with the attachment glycoprotein (G) develop lung eosinophilia after intranasal RSV challenge. Mucosal peptide vaccination reduced pulmonary eosinophilia in mice subsequently immunized with G and challenged with RSV. These studies emphasize that protective and immunoregulatory CD8(+) CTL responses can be mucosally elicited using enterotoxin-based mucosal adjuvants but that resistance against viral infection may be accompanied by enhanced disease.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Toxinas Bacterianas/imunologia , Enterotoxinas/imunologia , Proteínas de Escherichia coli , Mucosa Nasal/imunologia , Fragmentos de Peptídeos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais/imunologia , Administração Intranasal , Animais , Toxinas Bacterianas/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Enterotoxinas/administração & dosagem , Eosinofilia/imunologia , Eosinofilia/prevenção & controle , Epitopos de Linfócito T/imunologia , Imunidade Inata , Interferon gama/fisiologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Suínos , Linfócitos T Citotóxicos/virologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia , Proteínas Virais/administração & dosagem , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
18.
J Immunol ; 165(12): 7109-15, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11120841

RESUMO

Bronchiolitis caused by respiratory syncytial virus (RSV) infection is a major cause of hospitalization in children under 1 year of age. RSV causes common colds in older children and adults, but can cause serious disease in immunodeficient patients and the elderly. Development of effective vaccines and treatments for RSV infection is therefore a priority. Because bronchiolitis and vaccine-augmented disease are thought to be caused by exuberant T cell activation, attention has focused on the use of immunomodulators that affect T cell responses. In mice, IL-12 treatment down-regulates type 2 cytokine responses to the attachment protein G of RSV, reducing lung eosinophilia but further enhancing illness. We now show that CD8(+) T cells are responsible for enhanced weight loss, whereas IL-12-activated NK cells express high levels of IFN-gamma and inhibit lung eosinophilia without causing illness. Moreover, unlike immunocompetent mice, virus is detected in the mediastinal lymph nodes after elimination of both CD8(+) T cells and NK cells. These studies show that innate immune responses to viral infections direct the pattern of subsequent specific immunity and are critical to the development of nonpathogenic antiviral effects. We speculate that IL-12 treatment might be beneficial and safe in T cell-deficient patients with RSV pneumonitis.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteína HN/fisiologia , Interleucina-12/fisiologia , Células Matadoras Naturais/imunologia , Linfopenia/imunologia , Eosinofilia Pulmonar/imunologia , Eosinofilia Pulmonar/prevenção & controle , Vírus Sinciciais Respiratórios/fisiologia , Proteínas Virais/fisiologia , Animais , Antivirais/farmacologia , Linfócitos T CD8-Positivos/patologia , Movimento Celular/imunologia , Citocinas/biossíntese , Injeções Intraperitoneais , Interferon gama/farmacologia , Interferon gama/fisiologia , Interleucina-12/administração & dosagem , Ativação Linfocitária/imunologia , Linfopenia/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Eosinofilia Pulmonar/fisiopatologia , Eosinofilia Pulmonar/virologia , Índice de Gravidade de Doença , Subpopulações de Linfócitos T/imunologia , Proteínas do Envelope Viral
20.
J Exp Med ; 192(9): 1317-26, 2000 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-11067880

RESUMO

The effect of infection history is ignored in most animal models of infectious disease. The attachment protein of respiratory syncytial virus (RSV) induces T helper cell type 2-driven pulmonary eosinophilia in mice similar to that seen in the failed infant vaccinations in the 1960s. We show that previous influenza virus infection of mice: (a) protects against weight loss, illness, and lung eosinophilia; (b) attenuates recruitment of inflammatory cells; and (c) reduces cytokine secretion caused by RSV attachment protein without affecting RSV clearance. This protective effect can be transferred via influenza-immune splenocytes to naive mice and is long lived. Previous immunity to lung infection clearly plays an important and underestimated role in subsequent vaccination and infection. The data have important implications for the timing of vaccinations in certain patient groups, and may contribute to variability in disease susceptibility observed in humans.


Assuntos
Pulmão/imunologia , Pulmão/virologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Transplante de Células , Citocinas/metabolismo , Citotoxicidade Imunológica/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Eosinofilia/complicações , Eosinofilia/imunologia , Eosinofilia/patologia , Eosinofilia/virologia , Feminino , Proteína HN/imunologia , Histocitoquímica , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Orthomyxoviridae/imunologia , Orthomyxoviridae/fisiologia , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Infecções por Vírus Respiratório Sincicial/complicações , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/patogenicidade , Baço/citologia , Baço/imunologia , Superinfecção/imunologia , Superinfecção/patologia , Superinfecção/fisiopatologia , Superinfecção/virologia , Proteínas do Envelope Viral , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...