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










Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 16(4): 399-407, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37088262

RESUMO

The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. The airways have been linked with the trafficking of myelin-specific T-cells in the preclinical stages of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Th17 cells are important pathogenic effectors in MS and EAE but are innocuous immediately following differentiation. Upregulation of the cytokine GM-CSF appears to be a critical step in their acquisition of pathogenic potential, but little is known about the mechanisms that mediate this process. Here, primed myelin-specific Th17 cells were transferred to congenic recipient mice prior to exposure to various human respiratory tract-associated bacteria and T-cell trafficking, phenotype and the severity of resulting EAE were monitored. Disease was exacerbated in mice exposed to the Proteobacteria Moraxella catarrhalis and Klebsiella pneumoniae, but not the Firmicute Veillonella parvula, and this was associated with significantly increased GM-CSF+ and GM-CSF+IFNγ+ ex-Th17-like donor CD4 T cells in the lungs and central nervous system (CNS) of these mice. These findings support the concept that respiratory bacteria may contribute to the pathophysiology of CNS autoimmunity by modulating pathogenicity in crucial T-cell subsets that orchestrate neuroinflammation.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Camundongos , Humanos , Animais , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Moraxella catarrhalis , Klebsiella pneumoniae , Bainha de Mielina/patologia , Células Th17 , Virulência , Encefalomielite Autoimune Experimental/patologia , Esclerose Múltipla/patologia , Sistema Respiratório , Camundongos Endogâmicos C57BL , Células Th1
2.
Eur J Immunol ; 53(5): e2250247, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36681765

RESUMO

Tissue-resident memory CD4 T (TRM ) cells induced by infection with Bordetella pertussis persist in respiratory tissues and confer long-term protective immunity against reinfection. However, it is not clear how they are maintained in respiratory tissues. Here, we demonstrate that B. pertussis-specific CD4 TRM cells produce IL-17A in response to in vitro stimulation with LPS or heat-killed Klebsiella pneumoniae (HKKP) in the presence of dendritic cells. Furthermore, IL-17A-secreting CD4 TRM cells expand in the lung and nasal tissue of B. pertussis convalescent mice following in vivo administration of LPS or HKKP. Bystander activation of CD4 TRM cells was suppressed by anti-IL-12p40 but not by anti-MHCII antibodies. Furthermore, purified respiratory tissue-resident, but not circulating, CD4 T cells from convalescent mice produced IL-17A following direct stimulation with IL-23 and IL-1ß or IL-18. Intranasal immunization of mice with a whole-cell pertussis vaccine induced respiratory CD4 TRM cells that were reactivated following stimulation with K. pneumoniae. Furthermore, the nasal pertussis vaccine conferred protective immunity against B. pertussis but also attenuated infection with K. pneumoniae. Our findings demonstrate that CD4 TRM cells induced by respiratory infection or vaccination can undergo bystander activation and confer heterologous immunity to an unrelated respiratory pathogen.


Assuntos
Bordetella pertussis , Coqueluche , Animais , Camundongos , Bordetella pertussis/fisiologia , Coqueluche/prevenção & controle , Linfócitos T CD4-Positivos , Interleucina-17 , Klebsiella pneumoniae , Imunidade Heteróloga , Lipopolissacarídeos , Memória Imunológica , Vacina contra Coqueluche
3.
Clin Rev Allergy Immunol ; 64(2): 161-178, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35275333

RESUMO

The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. Increasing reports have linked changes in this microbiome to a range of pulmonary and extrapulmonary disorders, including asthma, chronic obstructive pulmonary disease and rheumatoid arthritis. Central to many of these findings is the role of IL-17-type immunity as an important driver of inflammation. Despite the crucial role played by IL-17-mediated immune responses in protection against infection, overt Th17 cell responses have been implicated in the pathogenesis of several chronic inflammatory diseases. However, our knowledge of the influence of bacteria that commonly colonise the respiratory tract on IL-17-driven inflammatory responses remains sparse. In this article, we review the current knowledge on the role of specific members of the airway microbiota in the modulation of IL-17-type immunity and discuss how this line of research may support the testing of susceptible individuals and targeting of inflammation at its earliest stages in the hope of preventing the development of chronic disease.


Assuntos
Inflamação , Interleucina-17 , Pulmão , Microbiota , Humanos , Doença Crônica , Inflamação/imunologia , Interleucina-17/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Microbiota/imunologia , Sistema Respiratório/imunologia , Sistema Respiratório/microbiologia
4.
J Immunol ; 205(4): 1039-1049, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32651220

RESUMO

Murine studies have shown the potential for γδ T cells to mediate immunity to Staphylococcus aureus in multiple tissue settings by the secretion of diverse cytokines. However, the role played by γδ T cells in human immune responses to S. aureus is almost entirely unknown. In this study, we establish the capacity of human Vδ2+ γδ T cells for rapid activation in response to S. aureus In coculture with S. aureus-infected monocyte-derived dendritic cells (DCs), Vδ2+ cells derived from peripheral blood rapidly upregulate CD69 and secrete high levels of IFN-γ. DCs mediate this response through direct contact and IL-12 secretion. In turn, IFN-γ released by Vδ2+ cells upregulates IL-12 secretion by DCs in a positive feedback loop. Furthermore, coculture with γδ T cells results in heightened expression of the costimulatory molecule CD86 and the lymph node homing molecule CCR7 on S. aureus-infected DCs. In cocultures of CD4+ T cells with S. aureus-infected DCs, the addition of γδ T cells results in heightened CD4+ T cell activation. Our findings identify γδ T cells as potential key players in the early host response to S. aureus during bloodstream infection, promoting enhanced responses by both innate and adaptive immune cell populations, and support their consideration in the development of host-directed anti-S. aureus treatments.


Assuntos
Imunidade Adaptativa/imunologia , Linfócitos T CD4-Positivos/imunologia , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígeno B7-2/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Humanos , Interferon gama/imunologia , Interleucina-12/imunologia , Lectinas Tipo C/imunologia , Monócitos/imunologia , Receptores CCR7/imunologia , Regulação para Cima/imunologia
5.
J Exp Med ; 217(5)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32106283

RESUMO

T cells are classically recognized as distinct subsets that express αß or γδ TCRs. We identify a novel population of T cells that coexpress αß and γδ TCRs in mice and humans. These hybrid αß-γδ T cells arose in the murine fetal thymus by day 16 of ontogeny, underwent αß TCR-mediated positive selection into CD4+ or CD8+ thymocytes, and constituted up to 10% of TCRδ+ cells in lymphoid organs. They expressed high levels of IL-1R1 and IL-23R and secreted IFN-γ, IL-17, and GM-CSF in response to canonically restricted peptide antigens or stimulation with IL-1ß and IL-23. Hybrid αß-γδ T cells were transcriptomically distinct from conventional γδ T cells and displayed a hyperinflammatory phenotype enriched for chemokine receptors and homing molecules that facilitate migration to sites of inflammation. These proinflammatory T cells promoted bacterial clearance after infection with Staphylococcus aureus and, by licensing encephalitogenic Th17 cells, played a key role in the development of autoimmune disease in the central nervous system.


Assuntos
Inflamação/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/imunologia , Animais , Biomarcadores/metabolismo , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Inflamação/patologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Fenótipo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T gama-delta/genética , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/fisiologia , Transcrição Gênica , Transcriptoma/genética
6.
Infect Immun ; 85(12)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28947645

RESUMO

Staphylococcus aureus has become increasingly resistant to antibiotics, and vaccines offer a potential solution to this epidemic of antimicrobial resistance. Targeting of specific T cell subsets is now considered crucial for next-generation anti-S. aureus vaccines; however, there is a paucity of information regarding T cell antigens of S. aureus This study highlights the importance of cell wall-anchored proteins as human CD4+ T cell activators capable of driving antigen-specific Th1 and Th17 cell activation. Clumping factor A (ClfA), which contains N1, N2, and N3 binding domains, was found to be a potent human T cell activator. We further investigated which subdomains of ClfA were involved in T cell activation and found that the full-length ClfA N123 and N23 were potent Th1 and Th17 activators. Interestingly, the N1 subdomain was capable of exclusively activating Th1 cells. Furthermore, when these subdomains were used in a model vaccine, N23 and N1 offered Th1- and Th17-mediated systemic protection in mice upon intraperitoneal challenge. Overall, however, full-length ClfA N123 is required for maximal protection both locally and systemically.


Assuntos
Antígenos de Bactérias/imunologia , Coagulase/imunologia , Staphylococcus aureus/imunologia , Linfócitos T/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/administração & dosagem , Vacinas Antiestafilocócicas/imunologia , Análise de Sobrevida
7.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L855-L860, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28336811

RESUMO

Our body clock drives rhythms in the expression of genes that have a 24-h periodicity. The transcription factor BMAL1 is a crucial component of the molecular clock. A number of physiological processes, including immune function, are modulated by the circadian clock. Asthma, a disease with very strong clinical evidence demonstrating regulation by circadian variation, is of particular relevance to circadian control of immunity. Airway hypersensitivity and asthma attacks are more common at night in humans. The molecular basis for this is unknown, and there is no model of asthma in animals with genetic distortion of the molecular clock. We used mice lacking BMAL1 in myeloid cells (BMAL1-LysM-/-) to determine the role of BMAL1 in allergic asthma. Using the ovalbumin model of allergic asthma, we demonstrated markedly increased asthma features, such as increased lung inflammation, demonstrated by drastically higher numbers of eosinophils and increased IL-5 levels in the lung and serum, in BMAL1-LysM-/- mice. In vitro studies demonstrated increased proinflammatory chemokine and mannose receptor expression in IL-4- as well as LPS-treated macrophages from BMAL1-LysM-/- mice compared with wild-type controls. This suggests that Bmal1 is a potent negative regulator in myeloid cells in the context of allergic asthma. Our findings might explain the increase in asthma incidents during the night, when BMAL1 expression is low.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Asma/complicações , Asma/metabolismo , Ritmo Circadiano , Hipersensibilidade/complicações , Hipersensibilidade/metabolismo , Células Mieloides/metabolismo , Animais , Asma/patologia , Biomarcadores/metabolismo , Quimiocinas/metabolismo , Modelos Animais de Doenças , Eosinófilos/metabolismo , Eosinófilos/patologia , Hipersensibilidade/patologia , Mediadores da Inflamação/metabolismo , Interleucina-5/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Pneumonia/complicações , Pneumonia/metabolismo , Pneumonia/patologia
8.
Trends Immunol ; 37(10): 690-702, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27567182

RESUMO

Despite the potential for diversity in their T cell receptor, γδ T cells are primarily considered to be innate immune cells. Recently, memory-like γδ T cell responses have been identified in murine models of infection and autoimmunity. Similar memory responses have also been described in human and non-human primate γδ T cells. It has thus become clear that subpopulations of γδ T cells can develop long-lasting memory akin to conventional αß T cells, with protective and pathogenic consequences. Hence, a re-evaluation of their true capabilities and role in infection and immunity is required. This review discusses recent reports of memory-type responses attributed to γδ T cells and assesses this underappreciated facet of these enigmatic cells.


Assuntos
Doenças Autoimunes/imunologia , Memória Imunológica , Infecções/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Humanos , Imunidade Inata , Camundongos , Primatas , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo
9.
J Neuroinflammation ; 13(1): 178, 2016 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-27391474

RESUMO

BACKGROUND: Neurodegeneration is believed to be the primary cause of permanent, long-term disability in patients with multiple sclerosis. The cause of neurodegeneration in multiple sclerosis appears to be multifactorial. One mechanism that has been implicated in the pathogenesis of neurodegeneration in multiple sclerosis is the targeting of neuronal and axonal antigens by autoantibodies. Multiple sclerosis patients develop antibodies to the RNA-binding protein, heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1), which is enriched in neurons. We hypothesized that anti-hnRNP A1 antibodies would contribute to neurodegeneration in an animal model of multiple sclerosis. METHODS: Following induction of experimental autoimmune encephalomyelitis (EAE) by direct immunization with myelin oligodendrocyte glycoprotein, mice were injected with anti-hnRNP A1 or control antibodies. Animals were examined clinically, and the central nervous system (CNS) tissues were tested for neurodegeneration with Fluoro-Jade C, a marker of degenerating neural elements. RESULTS: Injection of anti-hnRNP A1 antibodies in mice with EAE worsened clinical disease, altered the clinical disease phenotype, and caused neurodegeneration preferentially in the ventral spinocerebellar tract and deep white matter of the cerebellum in the CNS. Neurodegeneration in mice injected with hnRNP A1-M9 antibodies compared to control groups was consistent with "dying back" axonal degeneration. CONCLUSIONS: These data suggest that antibodies to the RNA-binding protein hnRNP A1 contribute to neurodegeneration in immune-mediated disease of the CNS.


Assuntos
Autoanticorpos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/imunologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Animais , Autoanticorpos/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/metabolismo
10.
PLoS Pathog ; 11(11): e1005226, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26539822

RESUMO

Mechanisms of protective immunity to Staphylococcus aureus infection in humans remain elusive. While the importance of cellular immunity has been shown in mice, T cell responses in humans have not been characterised. Using a murine model of recurrent S. aureus peritonitis, we demonstrated that prior exposure to S. aureus enhanced IFNγ responses upon subsequent infection, while adoptive transfer of S. aureus antigen-specific Th1 cells was protective in naïve mice. Translating these findings, we found that S. aureus antigen-specific Th1 cells were also significantly expanded during human S. aureus bloodstream infection (BSI). These Th1 cells were CD45RO+, indicative of a memory phenotype. Thus, exposure to S. aureus induces memory Th1 cells in mice and humans, identifying Th1 cells as potential S. aureus vaccine targets. Consequently, we developed a model vaccine comprising staphylococcal clumping factor A, which we demonstrate to be an effective human T cell antigen, combined with the Th1-driving adjuvant CpG. This novel Th1-inducing vaccine conferred significant protection during S. aureus infection in mice. This study notably advances our understanding of S. aureus cellular immunity, and demonstrates for the first time that a correlate of S. aureus protective immunity identified in mice may be relevant in humans.


Assuntos
Memória Imunológica , Infecções Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Células Th1/imunologia , Adjuvantes Imunológicos/farmacologia , Transferência Adotiva , Adulto , Idoso , Animais , Antígenos/imunologia , Feminino , Humanos , Interleucina-17/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Infecções Cutâneas Estafilocócicas/imunologia , Células Th1/efeitos dos fármacos
12.
J Immunol ; 192(8): 3697-708, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24623128

RESUMO

The development of vaccines against Staphylococcus aureus has consistently failed in clinical trials, likely due to inefficient induction of cellular immunity. T cell-derived IL-17 is one of the few known correlates of antistaphylococcoal immunity, conferring protection against S. aureus infections through its ability to promote phagocytic cell effector functions. A comprehensive understanding of the discrete T cell subsets critical for site-specific IL-17-mediated bacterial clearance will therefore be necessary to inform the development of vaccines that efficiently target cellular immunity. In this study, we have identified a population of CD44+ CD27- memory γδ T cells, expanded upon infection of C57BL/6 mice with S. aureus, which produce high levels of IL-17 and mediate enhanced bacterial clearance upon reinfection with the bacterium. These cells are comprised largely of the Vγ4+ subset and accumulate at the site of infection subsequent to an initial Vγ1.1+ and Vγ2+ T cell response. Moreover, these Vγ4+ T cells are retained in the peritoneum and draining mediastinal lymph nodes for a prolonged period following bacterial clearance. In contrast to its critical requirement for γδ T cell activation during the primary infection, IL-1 signaling was dispensable for activation and expansion of memory γδ T cells upon re-exposure to S. aureus. Our findings demonstrate that a γδ T cell memory response can be induced upon exposure to S. aureus, in a fashion analogous to that associated with classical αß T cells, and suggest that induction of IL-17-expressing γδ T cells may be an important property of a protective vaccine against S. aureus.


Assuntos
Memória Imunológica , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transferência Adotiva , Animais , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Imunidade Inata , Interleucina-17/biossíntese , Interleucina-17/genética , Masculino , Camundongos , Camundongos Knockout , Peritonite/imunologia , Peritonite/microbiologia , Transdução de Sinais , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/terapia
13.
Eur J Immunol ; 43(11): 2866-74, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23873018

RESUMO

Drugs that block leukocyte trafficking ameliorate multiple sclerosis (MS). Occurrences of opportunistic infection, however, highlight the need for novel drugs that modulate more restricted subsets of T cells. In this context, chemokines and their receptors are attractive therapeutic targets. CXCR3, a Th1-associated chemokine receptor, is preferentially expressed on T cells that accumulate in MS lesions and central nervous system (CNS) infiltrates of mice with experimental autoimmune encephalomyelitis (EAE). Surprisingly, mice genetically deficient in either CXCR3 or CXCL10 succumb to EAE following active immunization with myelin antigens. EAE is mediated by a heterogeneous population of T cells in myelin-immunized mice. Hence, disease might develop in the absence of CXCR3 secondary to the compensatory action of encephalitogenic CCR6(+) Th17 cells. However, in the current study, we show for the first time that blockade or genetic deficiency of either CXCR3 or of its primary ligand has no impact on clinical EAE induced by the adoptive transfer of highly polarized Th1 effector cells. Our data illustrate the fact that, although highly targeted immunotherapies might have more favorable side effect profiles, they are also more likely to be rendered ineffective by inherent redundancies in chemokine and cytokine networks that arise at sites of neuroinflammation.


Assuntos
Quimiocina CXCL10/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Receptores CXCR3/metabolismo , Células Th1/imunologia , Transferência Adotiva , Animais , Sistema Nervoso Central/citologia , Quimiocina CXCL10/deficiência , Quimiocina CXCL10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/imunologia , Receptores CXCR3/genética , Transdução de Sinais , Células Th1/transplante
14.
Eur J Immunol ; 43(11): 2824-31, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23878008

RESUMO

In the MOG35-55 induced EAE model, autoreactive Th17 cells that accumulate in the central nervous system acquire Th1 characteristics via a T-bet dependent mechanism. It remains to be determined whether Th17 plasticity and encephalitogenicity are causally related to each other. Here, we show that IL-23 polarized T-bet(-/-) Th17 cells are unimpaired in either activation or proliferation, and induce higher quantities of the chemokines RANTES and CXCL2 than WT Th17 cells. Unlike their WT counterparts, T-bet(-/-) Th17 cells retain an IL-17(hi) IFN-γ(neg-lo) cytokine profile following adoptive transfer into syngeneic hosts. This population of highly polarized Th17 effectors is capable of mediating EAE, albeit with a milder clinical course. It has previously been reported that the signature Th1 and Th17 effector cytokines, IFN-γ and IL-17, are dispensable for the development of autoimmune demyelinating disease. The current study demonstrates that the "master regulator" transcription factor, T-bet, is also not universally required for encephalitogenicity. Our results contribute to a growing body of data showing heterogeneity of myelin-reactive T cells and the independent mechanisms they employ to inflict damage to central nervous system tissues, complicating the search for therapeutic targets relevant across the spectrum of individuals with multiple sclerosis.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Proteínas com Domínio T/metabolismo , Células Th17/metabolismo , Animais , Diferenciação Celular/imunologia , Polaridade Celular/imunologia , Proliferação de Células , Sistema Nervoso Central/imunologia , Quimiocina CCL5/biossíntese , Quimiocina CXCL2/biossíntese , Proteínas de Ligação a DNA/genética , Encefalomielite Autoimune Experimental/genética , Interferon gama/biossíntese , Interleucina-17/biossíntese , Interleucina-23 , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas com Domínio T/genética , Células Th1/imunologia , Células Th17/imunologia
15.
J Immunol ; 186(10): 5738-48, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21471445

RESUMO

IL-1ß plays a critical role in promoting IL-17 production by γδ and CD4 T cells. However, IL-1-targeted drugs, although effective against autoinflammatory diseases, are less effective against autoimmune diseases. Conversely, gain-of-function mutations in the NLRP3 inflammasome complex are associated with enhanced IL-1ß and IL-18 production and Th17 responses. In this study, we examined the role of caspase-1-processed cytokines in IL-17 production and in induction of experimental autoimmune encephalomyelitis (EAE). Killed Mycobacterium tuberculosis, the immunostimulatory component in CFA used for inducing EAE, stimulated IL-1ß and IL-18 production by dendritic cells through activation of the inflammasome complex and caspase-1. Dendritic cells stimulated with M. tuberculosis and myelin oligodendrocyte glycoprotein promoted IL-17 production by T cells and induced EAE following transfer to naive mice, and this was suppressed by a caspase-1 inhibitor and reversed by administration of IL-1ß or IL-18. Direct injection of the caspase-1 inhibitor suppressed IL-17 production by CD4 T cells and γδ T cells in vivo and attenuated the clinical signs of EAE. γδ T cells expressed high levels of IL-18R and the combination of IL-18 and IL-23, as with IL-1ß and IL-23, stimulated IL-17 production by γδ T cells, but also from CD4 T cells, in the absence of TCR engagement. Our findings demonstrate that caspase-1-processed cytokines IL-1ß and IL-18 not only promote autoimmunity by stimulating innate IL-17 production by T cells but also reveal redundancy in the functions of IL-1ß and IL-18, suggesting that caspase-1 or the inflammasome may be an important drug target for autoimmune diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Caspase 1/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Interleucina-17/biossíntese , Interleucina-18/imunologia , Interleucina-1beta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Animais , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/metabolismo , Proteínas de Transporte/genética , Inibidores de Caspase , Células Dendríticas/imunologia , Interleucina-17/imunologia , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Interleucina-23/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Mycobacterium tuberculosis/imunologia , Proteínas da Mielina , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Proteína 3 que Contém Domínio de Pirina da Família NLR , Reação em Cadeia da Polimerase , Receptores de Antígenos de Linfócitos T gama-delta/genética , Células Th17/imunologia
16.
Brain Behav Immun ; 25(5): 922-31, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20933590

RESUMO

Cases of progressive multifocal leukoencephalopathy can occur in patients treated with the B cell depleting anti-CD20 antibody, rituximab, highlighting the importance of B cell surveillance of the central nervous system (CNS). The lymphoid chemokine, CXCL13, is critical for B cell recruitment and functional organization of peripheral lymphoid tissues, and CXCL13 levels are often elevated in the inflamed CNS. To more directly investigate the role of CXCL13 in CNS B cell migration, its role in animal models of infectious and inflammatory demyelinating disease was examined. During acute alphavirus encephalitis where viral clearance depends on the local actions of anti-viral antibodies, CXCL13 levels and B cell numbers increased in brain tissue over time. Surprisingly, however, CXCL13-deficient animals showed normal CNS B cell recruitment, unaltered CNS virus replication and clearance, and intact peripheral anti-viral antibody responses. During experimental autoimmune encephalomyelitis (EAE), CNS levels of CXCL13 increased as symptoms emerged and equivalent numbers of B cells were identified among the CNS infiltrates of CXCL13-deficient mice compared to control animals. However, CXCL13-deficient mice did not sustain pathogenic anti-myelin T cell responses, consistent with their known propensity to develop more self-limited EAE. These data show that CXCL13 is dispensable for CNS B cell recruitment in both models. The disease course is unaffected by CXCL13 in a CNS infection paradigm that depends on a pathogen-specific B cell response, while it is heightened and prolonged by CXCL13 when myelin-specific CD4+ T cells drive CNS pathology. Thus, CXCL13 could be a therapeutic target in certain neuroinflammatory diseases, but not by blocking B cell recruitment to the CNS.


Assuntos
Infecções por Alphavirus/imunologia , Linfócitos B/fisiologia , Quimiocina CXCL13/fisiologia , Quimiotaxia de Leucócito/fisiologia , Encefalite Viral/imunologia , Encefalomielite Autoimune Experimental/imunologia , Vírus Sindbis/imunologia , Infecções por Alphavirus/fisiopatologia , Animais , Astrócitos/imunologia , Astrócitos/virologia , Encéfalo/imunologia , Encéfalo/virologia , Células Cultivadas , Encefalite Viral/fisiopatologia , Encefalomielite Autoimune Experimental/fisiopatologia , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/virologia , Reação em Cadeia da Polimerase , Células Th1/imunologia , Células Th1/fisiologia , Células Th17/imunologia , Células Th17/fisiologia
17.
J Neuroimmunol ; 224(1-2): 56-61, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20605642

RESUMO

Lymphoid chemokines, including CCL19, CCL21 and CXCL13, are critical in the development and organization of secondary lymphoid tissues and in the generation of adaptive immune responses. These molecules have also been implicated in the development of ectopic lymphoid structures in the setting of chronic inflammation. Here we review current knowledge on the production of lymphoid chemokines in the central nervous system during both homeostatic conditions and in disease states. Accumulating evidence suggests that constitutive expression of CCL19 plays a critical immunosurveillance role in healthy individuals. In contrast, aberrant induction of CCL19, CCL21 and CXCL13 may support the establishment of chronic autoimmunity and hematopoietic tumors within the CNS.


Assuntos
Doenças do Sistema Nervoso Central/imunologia , Doenças do Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Quimiocinas/fisiologia , Linfócitos/imunologia , Linfócitos/metabolismo , Animais , Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/patologia , Quimiocinas/biossíntese , Homeostase/imunologia , Humanos , Mediadores da Inflamação/fisiologia , Linfócitos/patologia
18.
Brain Behav Immun ; 24(4): 641-51, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20138983

RESUMO

Experimental autoimmune encephalomyelitis (EAE) is a mouse model for multiple sclerosis, where disease is mediated by autoantigen-specific T cells. Although there is evidence linking CD4(+) T cells that secrete IL-17, termed Th17 cells, and IFN-gamma-secreting Th1 cells with the pathogenesis of EAE, the precise contribution of these T cell subtypes or their associated cytokines is still unclear. We have investigated the infiltration of CD4(+) T cells that secrete IFN-gamma, IL-17 or both cytokines into CNS during development of EAE and have examined the role of T cells in microglial activation. Our findings demonstrate that Th17 cells and CD4(+) T cells that produce both IFN-gamma and IL-17, which we have called Th1/Th17 cells, infiltrate the brain prior to the development of clinical symptoms of EAE and that this coincides with activation of CD11b(+) microglia and local production of IL-1beta, TNF-alpha and IL-6 in the CNS. In contrast, significant infiltration of Th1 cells was only detected after the development of clinical disease. Co-culture experiments, using mixed glia and MOG-specific T cells, revealed that T cells that secreted IFN-gamma and IL-17 were potent activators of pro-inflammatory cytokines but T cells that secrete IFN-gamma, but not IL-17, were less effective. In contrast both Th1 and Th1/Th17 cells enhanced MHC-class II and co-stimulatory molecule expression on microglia. Our findings suggest that T cells which secrete IL-17 or IL-17 and IFN-gamma infiltrate the CNS prior to the onset of clinical symptoms of EAE, where they may mediate CNS inflammation, in part, through microglial activation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Microglia/imunologia , Animais , Antígeno CD11b/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Sistema Nervoso Central/citologia , Técnicas de Cocultura , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Ensaio de Imunoadsorção Enzimática , Glicoproteínas , Injeções Subcutâneas , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos , Reação em Cadeia da Polimerase , Células Th1/imunologia , Células Th1/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
19.
Immunity ; 31(2): 331-41, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19682929

RESUMO

Th17 cells, CD4(+) T cells that secrete interleukin-17 (IL-17), are pathogenic in autoimmune diseases and their development and expansion is driven by the cytokines IL-6, TGF-beta, IL-21, IL-1, and IL-23. However, there are also innate sources of IL-17. Here, we show that gammadelta T cells express IL-23R and the transcription factor RORgammat and produce IL-17, IL-21, and IL-22 in response to IL-1beta and IL-23, without T cell receptor engagement. IL-17-producing gammadelta T cells were found at high frequency in the brain of mice with experimental autoimmune encephalomyelitis (EAE). gammadelta T cells activated by IL-1beta and IL-23 promoted IL-17 production by CD4(+) T cells and increased susceptibility to EAE, suggesting that gammadelta T cells act in an amplification loop for IL-17 production by Th17 cells. Our findings demonstrate that gammadelta T cells activated by IL-1beta and IL-23 are an important source of innate IL-17 and IL-21 and provide an alternative mechanism whereby IL-1 and IL-23 may mediate autoimmune inflammation.


Assuntos
Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Interleucina-17/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Interleucina-17/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Autoimunidade , Complexo CD3/imunologia , Complexo CD3/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Interleucina-17/biossíntese , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacologia , Interleucina-23/imunologia , Interleucina-23/metabolismo , Interleucina-23/farmacologia , Interleucinas/imunologia , Interleucinas/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Interleucina/imunologia , Receptores de Interleucina/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-17/imunologia , Receptores do Ácido Retinoico/imunologia , Receptores do Ácido Retinoico/metabolismo , Receptores dos Hormônios Tireóideos/imunologia , Receptores dos Hormônios Tireóideos/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/metabolismo
20.
J Immunol ; 183(3): 1715-23, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19570828

RESUMO

IL-17-producing CD4(+) T (Th17) cells are pathogenic in many autoimmune diseases. The induction and expansion of Th17 cells is directed by cytokines, including IL-23 and IL-1beta, produced by innate immune cells through activation of pathogen recognition receptors. The NF-kappaB and IFN regulatory factor families of transcriptional factors mediate IL-12 production; however, distinct signaling pathways appear to be required for IL-23 production. In this study, we show that inhibition of ERK MAPK suppressed IL-23 and IL-1beta production by dendritic cells stimulated with TLR or dectin-1 agonists but did not affect IL-12p70 production. Furthermore, an ERK inhibitor suppressed the ability of Ag-pulsed TLR-activated dendritic cells to induce Ag-specific Th17 cells in vivo, but interestingly also inhibited the induction of Th1 cells. Treatment with an ERK inhibitor attenuated experimental autoimmune encephalomyelitis (EAE), when administered either at the induction phase of acute EAE or during remission in the relapsing-remitting EAE model. This was associated with significant suppression of autoantigen-specific Th17 and Th1 responses. The suppressive effect of the ERK inhibitor on attenuation of EAE was reversed by administration of IL-1beta and IL-23. Our findings suggest that ERK MAPK plays a critical and hitherto undescribed role in activating innate production of IL-23 and IL-1beta, which promote pathogenic T cell responses, and therefore represents an important target for therapeutic intervention against autoimmune diseases.


Assuntos
Doenças Autoimunes/imunologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Interleucina-17/biossíntese , Interleucina-1beta/imunologia , Interleucina-23/imunologia , Animais , Células Cultivadas , Células Dendríticas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Interleucina-1beta/biossíntese , Interleucina-23/biossíntese , Camundongos , Linfócitos T/imunologia , Células Th1 , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...