Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(7): e1011460, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37405965

RESUMO

Recruiting large numbers of naïve lymphocytes to lymph nodes is critical for mounting an effective adaptive immune response. While most naïve lymphocytes utilize homing molecule L-selectin to enter lymph nodes, some circulating cells can traffic to the lung-draining mediastinal lymph node (mLN) through lymphatics via the intermediate organ, lung. However, whether this alternative trafficking mechanism operates in infection and contributes to T cell priming are unknown. We report that in pulmonary Mycobacterium tuberculosis-infected mice, homing of circulating lymphocytes to the mLN is significantly less efficient than to non-draining lymph node. CD62L blockade only partially reduced the homing of naïve T lymphocytes, consistent with L-selectin-independent routing of naïve lymphocytes to the site. We further demonstrated that lymphatic vessels in infected mLN expanded significantly and inhibiting lymphangiogenesis with a vascular endothelial growth factor receptor 3 kinase inhibitor reduced the recruitment of intravenously injected naïve lymphocytes to the mLN. Finally, mycobacterium-specific T cells entering via the L-selectin-independent route were readily activated in the mLN. Our study suggests that both L-selectin-dependent and -independent pathways contribute to naïve lymphocyte entry into mLN during M. tuberculosis infection and the latter pathway may represent an important mechanism for orchestrating host defence in the lungs.


Assuntos
Mycobacterium tuberculosis , Tuberculose Pulmonar , Tuberculose , Camundongos , Animais , Selectina L/metabolismo , Linfócitos T , Fator A de Crescimento do Endotélio Vascular/metabolismo , Linfócitos , Pulmão , Linfonodos , Tuberculose/metabolismo
2.
J Immunol ; 208(11): 2515-2522, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35534212

RESUMO

The cannabinoid receptor 2 (CB2) is a receptor mainly expressed in immune cells and believed to be immunosuppressive in infective or inflammatory models. However, its role in sepsis has not been fully elucidated. In this study, we delineate the function and mechanism of CB2 in the cecal ligation and puncture-induced septic model in mice. The activation of CB2 signaling with HU308 led to decreased survival rates and more severe lung injury in septic mice, and lower IL-10 levels in peritoneal lavage fluid were observed in the CB2 agonist group. The mice with conditional knockout of CB2-encoding gene CNR2 in CD4+ T cells (CD4 Cre CNR2fl/fl) improved survival, enhanced IL-10 production, and ameliorated pulmonary damage in the sepsis model after CB2 activation. In addition, double-knockout of the CNR2 gene (Lyz2 Cre CD4 Cre CNR2fl/fl) decreased the susceptibility to sepsis compared with Lyz2 Cre CNR2fl/fl mice. Mechanistically, the blockade of IL-10 with the anti-IL-10 Ab abolished its protection in CD4 Cre CNR2fl/fl mice. In accordance with the animal study, in vitro results revealed that the lack of CNR2 in CD4+ cells elevated IL-10 production, and CB2 activation inhibited CD4+ T cell-derived IL-10 production. Furthermore, in the clinical environment, septic patients expressed enhanced CB2 mRNA levels compared with healthy donors in PBMCs, and their CB2 expression was inversely correlated with IL-10. These results suggested that the activation of CD4+ T cell-derived CB2 increased susceptibility to sepsis through inhibiting IL-10 production.


Assuntos
Linfócitos T CD4-Positivos , Interleucina-10 , Receptor CB2 de Canabinoide , Sepse , Animais , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Receptor CB2 de Canabinoide/genética , Sepse/patologia
3.
J Immunol ; 206(12): 2875-2887, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34049970

RESUMO

The quality of T cell responses depends on the lymphocytes' ability to undergo clonal expansion, acquire effector functions, and traffic to the site of infection. Although TCR signal strength is thought to dominantly shape the T cell response, by using TCR transgenic CD4+ T cells with different peptide:MHC binding affinity, we reveal that TCR affinity does not control Th1 effector function acquisition or the functional output of individual effectors following mycobacterial infection in mice. Rather, TCR affinity calibrates the rate of cell division to synchronize the distinct processes of T cell proliferation, differentiation, and trafficking. By timing cell division-dependent IL-12R expression, TCR affinity controls when T cells become receptive to Th1-imprinting IL-12 signals, determining the emergence and magnitude of the Th1 effector pool. These findings reveal a distinct yet cooperative role for IL-12 and TCR binding affinity in Th1 differentiation and suggest that the temporal activation of clones with different TCR affinity is a major strategy to coordinate immune surveillance against persistent pathogens.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Mycobacterium bovis/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
4.
J Infect Dis ; 225(5): 825-835, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32939551

RESUMO

Previous studies demonstrated that transforming growth factor (TGT) ß1 plays an immunosuppressive role in clinical tuberculosis. However, the contribution of TGF-ß1 gene polymorphisms to human tuberculosis susceptibility remains undetermined. In this study, we showed that single-nucleotide polymorphisms (SNPs) in TGF-ß1 gene were associated with increased susceptibility to tuberculosis in the discovery cohort (1533 case patients and 1445 controls) and the validation cohort (832 case patients and 1084 controls), and 2 SNPs located in the promoter region (rs2317130 and rs4803457) are in strong linkage disequilibrium. The SNP rs2317130 was associated with the severity of tuberculosis. Further investigation demonstrated that rs2317130 CC genotype is associated with higher TGF-ß1 and interleukin 17A production. The mechanistic study showed that rs2317130 C allele affected TGF-ß1 promoter activity by regulating binding activity to nuclear extracts. These findings provide insights into the pathogenic role of TGF-ß1 in human tuberculosis and reveal a function for the TGF-ß1 promoter SNPs in regulating immune responses during Mycobacterium tuberculosis infection.


Assuntos
Fator de Crescimento Transformador beta1 , Tuberculose , Humanos , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Polimorfismo de Nucleotídeo Único , Fator de Crescimento Transformador beta , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Tuberculose/genética
5.
PLoS Pathog ; 16(5): e1008553, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32453761

RESUMO

IRGM and its mouse orthologue Irgm1 are dynamin-like proteins that regulate vesicular remodeling, intracellular microbial killing, and pathogen immunity. IRGM dysfunction is linked to inflammatory bowel disease (IBD), and while it is thought that defective intracellular killing of microbes underscores IBD susceptibility, studies have yet to address how IRGM/Irgm1 regulates immunity to microbes relevant to intestinal inflammation. Here we find that loss of Irgm1 confers marked susceptibility to Citrobacter rodentium, a noninvasive intestinal pathogen that models inflammatory responses to intestinal bacteria. Irgm1-deficient mice fail to control C. rodentium outgrowth in the intestine, leading to systemic pathogen spread and host mortality. Surprisingly, susceptibility due to loss of Irgm1 function was not linked to defective intracellular killing of C. rodentium or exaggerated inflammation, but was instead linked to failure to remodel specific colon lamina propria (C-LP) myeloid cells that expand in response to C. rodentium infection and are essential for C. rodentium immunity. Defective immune remodeling was most striking in C-LP monocytes, which were successfully recruited to the infected C-LP, but subsequently underwent apoptosis. Apoptotic susceptibility was induced by C. rodentium infection and was specific to this setting of pathogen infection, and was not apparent in other settings of intestinal inflammation. These studies reveal a novel role for Irgm1 in host defense and suggest that deficiencies in survival and remodeling of C-LP myeloid cells that control inflammatory intestinal bacteria may underpin IBD pathogenesis linked to IRGM dysfunction.


Assuntos
Citrobacter rodentium/imunologia , Colo/imunologia , Infecções por Enterobacteriaceae/imunologia , Proteínas de Ligação ao GTP/deficiência , Doenças Inflamatórias Intestinais/imunologia , Monócitos/imunologia , Animais , Colo/microbiologia , Colo/patologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/patologia , Proteínas de Ligação ao GTP/imunologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Camundongos , Camundongos Knockout , Monócitos/microbiologia , Monócitos/patologia , Mucosa/imunologia , Mucosa/microbiologia , Mucosa/patologia
6.
Fish Shellfish Immunol ; 120: 429-433, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34922016

RESUMO

Pidotimod (PDT) is a synthetic dipeptide molecule which can improve immune responses in mice and humans, protecting hosts from infection. However, the exact mechanism of protection remains ill-defined. The effect of pidotimod has not yet been investigated in the inflammatory response of zebrafish. In this study, we used tail wound and infection models of zebrafish to study the effect of PDT on inflammation. We found that zebrafish larvae were sensitive to PDT immersion causing toxicity at doses above 50 µg/mL. The tail wound assay showed that PDT increased the recruitment of neutrophils and macrophages to the wound site and promoted the transcription of the pro-inflammatory cytokine il1b. However, we did not observe protection of uropathogenic Escherichia coli or Mycobacterium marinum infected zebrafish larvae following PDT treatment. This study provides a new platform for PDT research, which is worthy of further research to identify further effects of PDT therapy.


Assuntos
Inflamação , Ácido Pirrolidonocarboxílico/análogos & derivados , Tiazolidinas/efeitos adversos , Peixe-Zebra , Animais , Inflamação/induzido quimicamente , Larva , Ácido Pirrolidonocarboxílico/efeitos adversos
7.
Immunol Cell Biol ; 99(8): 894-906, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34080230

RESUMO

Regulatory T cells (Tregs) play a critical role in immune regulation and peripheral tolerance. While different types of Tregs have been identified in both mice and humans, much of our understanding about how these cells maintain immune homeostasis is derived from animal models. In this study, we examined two distinct human lymphoid organs to understand how repeated exposure to infections at the mucosal surface influences the phenotype and tissue localization of Tregs. We show that while Tregs in both tonsils and spleen express a tissue-resident phenotype, they accumulate in greater numbers in tonsils. Tonsillar-resident Tregs exhibit a highly suppressive phenotype with significantly increased expression of CD39, ICOS and CTLA-4 compared with their counterparts in circulation or in the spleen. Functionally, resident Tregs are able effectively to suppress T cell proliferation. We further demonstrate that tonsillar-resident Tregs share key features of T follicular helper cells. Spatial analysis reveals that the vast majority of resident Tregs are localized at the border of the T-zone and B cell follicle, as well as within the lymphocyte pockets enriched with resident memory T cells. Together our findings suggest that resident Tregs are strategically co-localized to maintain immune homeostasis at sites of recurrent inflammation.


Assuntos
Ativação Linfocitária , Linfócitos T Reguladores , Animais , Linfócitos B , Humanos , Camundongos , Fenótipo
8.
Nat Immunol ; 9(11): 1279-87, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18806793

RESUMO

Mice deficient in the interferon-gamma (IFN-gamma)-inducible, immunity-related GTPase Irgm1 have defective host resistance to a variety of intracellular pathogens. This greater susceptibility to infection is associated with impaired IFN-gamma-dependent macrophage microbicidal activity in vitro. Here we show that Irgm1 also regulated the survival of mature effector CD4(+) T lymphocytes by protecting them from IFN-gamma-induced autophagic cell death. Mice deficient in both IFN-gamma and Irgm1 were 'rescued' from the lymphocyte depletion and greater mortality that occurs in mice singly deficient in Irgm1 after mycobacterial infection. Our studies identify a feedback mechanism in the T helper type 1 response that limits the detrimental effects of IFN-gamma on effector T lymphocyte survival while promoting the antimicrobial functions of IFN-gamma.


Assuntos
Autofagia , Linfócitos T CD4-Positivos/imunologia , Proteínas de Ligação ao GTP/imunologia , Interferon gama/imunologia , Animais , Autofagia/genética , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/ultraestrutura , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Feminino , Proteínas de Ligação ao GTP/genética , Interferon gama/genética , Interferon gama/farmacologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium avium/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Tuberculose/imunologia
9.
Immunity ; 34(5): 807-19, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21596592

RESUMO

Cell-mediated adaptive immunity is critical for host defense, but little is known about T cell behavior during delivery of effector function. Here we investigate relationships among antigen presentation, T cell motility, and local production of effector cytokines by CD4+ T cells within hepatic granulomas triggered by Bacille Calmette-Guérin or Mycobacterium tuberculosis. At steady-state, only small fractions of mycobacteria-specific T cells showed antigen-induced migration arrest within granulomas, resulting in low-level, polarized secretion of cytokines. However, exogenous antigen elicited rapid arrest and robust cytokine production by the vast majority of effector T cells. These findings suggest that limited antigen presentation and/or recognition within granulomas evoke a muted T cell response drawing on only a fraction of the host's potential effector capacity. Our results provide new insights into the regulation of host-protective functions, especially how antigen availability influences T cell dynamics and, in turn, effector T cell function during chronic infection.


Assuntos
Apresentação de Antígeno , Granuloma/imunologia , Hepatopatias/imunologia , Mycobacterium bovis/imunologia , Mycobacterium tuberculosis/imunologia , Linfócitos T/imunologia , Animais , Movimento Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/imunologia , Granuloma/microbiologia , Hepatopatias/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/citologia
10.
Immunity ; 35(6): 1023-34, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22195750

RESUMO

Interleukin-1 (IL-1) receptor signaling is necessary for control of Mycobacterium tuberculosis (Mtb) infection, yet the role of its two ligands, IL-1α and IL-1ß, and their regulation in vivo are poorly understood. Here, we showed that both IL-1α and IL-1ß are critically required for host resistance and identified two multifunctional inflammatory monocyte-macrophage and DC populations that coexpressed both IL-1 species at the single-cell level in lungs of Mtb-infected mice. Moreover, we demonstrated that interferons (IFNs) played important roles in regulating IL-1 production by these cells in vivo. Type I interferons inhibited IL-1 production by both subsets whereas CD4(+) T cell-derived IFN-γ selectively suppressed monocyte-macrophages. These data provide a cellular basis for both the anti-inflammatory effects of IFNs and probacterial functions of type I IFNs during Mtb infection and reveal differential regulation of IL-1 production by distinct cell populations as an additional layer of complexity in the activity of IL-1 in vivo.


Assuntos
Interferons/metabolismo , Interleucina-1alfa/biossíntese , Interleucina-1beta/biossíntese , Pulmão/imunologia , Mycobacterium tuberculosis/imunologia , Células Mieloides/imunologia , Tuberculose Pulmonar/imunologia , Animais , Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Humanos , Subunidade p40 da Interleucina-12/biossíntese , Pulmão/metabolismo , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Fagócitos/imunologia , Fagócitos/metabolismo , Fagócitos/microbiologia , Transdução de Sinais
11.
PLoS Pathog ; 12(1): e1005378, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26731100

RESUMO

Host control of influenza A virus (IAV) is associated with exuberant pulmonary inflammation characterized by the influx of myeloid cells and production of proinflammatory cytokines including interferons (IFNs). It is unclear, however, how the immune system clears the virus without causing lethal immunopathology. Here, we demonstrate that in addition to its known anti-viral activity, STAT1 signaling coordinates host inflammation during IAV infection in mice. This regulatory mechanism is dependent on both type I IFN and IFN-γ receptor signaling and, importantly, requires the functional interplay between the two pathways. The protective function of type I IFNs is associated with not only the recruitment of classical inflammatory Ly6Chi monocytes into IAV-infected lungs, but also the prevention of excessive monocyte activation by IFN-γ. Unexpectedly, type I IFNs preferentially regulate IFN-γ signaling in Ly6Clo rather than inflammatory Ly6Chi mononuclear cell populations. In the absence of type I IFN signaling, Ly6Clo monocytes/macrophages, become phenotypically and functionally more proinflammatory than Ly6Chi cells, revealing an unanticipated function of the Ly6Clo mononuclear cell subset in tissue inflammation. In addition, we show that type I IFNs employ distinct mechanisms to regulate monocyte and neutrophil trafficking. Type I IFN signaling is necessary, but not sufficient, for preventing neutrophil recruitment into the lungs of IAV-infected mice. Instead, the cooperation of type I IFNs and lymphocyte-produced IFN-γ is required to regulate the tissue neutrophilic response to IAV. Our study demonstrates that IFN interplay links innate and adaptive anti-viral immunity to orchestrate tissue inflammation and reveals an additional level of complexity for IFN-dependent regulatory mechanisms that function to prevent excessive immunopathology while preserving anti-microbial functions.


Assuntos
Vírus da Influenza A/imunologia , Interferon Tipo I/imunologia , Interferon gama/imunologia , Infecções por Orthomyxoviridae/imunologia , Pneumonia/imunologia , Infecções Respiratórias/imunologia , Imunidade Adaptativa/imunologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Imunidade Inata/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia
12.
PLoS Pathog ; 12(8): e1005799, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27540722

RESUMO

Disruption of T cell memory during severe immune suppression results in reactivation of chronic viral infections, such as Epstein Barr virus (EBV) and Cytomegalovirus (CMV). How different subsets of memory T cells contribute to the protective immunity against these viruses remains poorly defined. In this study we examined the compartmentalization of virus-specific, tissue resident memory CD8+ T cells in human lymphoid organs. This revealed two distinct populations of memory CD8+ T cells, that were CD69+CD103+ and CD69+CD103-, and were retained within the spleen and tonsils in the absence of recent T cell stimulation. These two types of memory cells were distinct not only in their phenotype and transcriptional profile, but also in their anatomical localization within tonsils and spleen. The EBV-specific, but not CMV-specific, CD8+ memory T cells preferentially accumulated in the tonsils and acquired a phenotype that ensured their retention at the epithelial sites where EBV replicates. In vitro studies revealed that the cytokine IL-15 can potentiate the retention of circulating effector memory CD8+ T cells by down-regulating the expression of sphingosine-1-phosphate receptor, required for T cell exit from tissues, and its transcriptional activator, Kruppel-like factor 2 (KLF2). Within the tonsils the expression of IL-15 was detected in regions where CD8+ T cells localized, further supporting a role for this cytokine in T cell retention. Together this study provides evidence for the compartmentalization of distinct types of resident memory T cells that could contribute to the long-term protection against persisting viral infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Herpesvirus Humano 4/imunologia , Memória Imunológica , Antígenos CD/imunologia , Linfócitos T CD8-Positivos/patologia , Infecções por Citomegalovirus/patologia , Infecções por Vírus Epstein-Barr/patologia , Feminino , Humanos , Interleucina-15/imunologia , Fatores de Transcrição Kruppel-Like/imunologia , Masculino , Especificidade de Órgãos/imunologia
13.
J Immunol ; 194(6): 2455-65, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25747907

RESUMO

Type I IFNs are known to inhibit viral replication and mediate protection against viral infection. However, recent studies revealed that these cytokines play a broader and more fundamental role in host responses to infections beyond their well-established antiviral function. Type I IFN induction, often associated with microbial evasion mechanisms unique to virulent microorganisms, is now shown to increase host susceptibility to a diverse range of pathogens, including some viruses. This article presents an overview of the role of type I IFNs in infections with bacterial, fungal, parasitic, and viral pathogens and discusses the key mechanisms mediating the regulatory function of type I IFNs in pathogen clearance and tissue inflammation.


Assuntos
Imunidade/imunologia , Infecções/imunologia , Interferon Tipo I/imunologia , Transdução de Sinais/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Resistência à Doença/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Infecções/microbiologia , Infecções/parasitologia , Interferon Tipo I/metabolismo , Receptor de Interferon alfa e beta/imunologia , Receptor de Interferon alfa e beta/metabolismo
14.
PLoS Pathog ; 10(10): e1004426, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25329476

RESUMO

Mycobacterium tuberculosis infection is associated with a spectrum of clinical outcomes, from long-term latent infection to different manifestations of progressive disease. Pro-inflammatory pathways, such as those controlled by IL-1ß, have the contrasting potential both to prevent disease by restricting bacterial replication, and to promote disease by inflicting tissue damage. Thus, the ultimate contribution of individual inflammatory pathways to the outcome of M. tuberculosis infection remains ambiguous. In this study, we identified a naturally-occurring polymorphism in the human IL1B promoter region, which alters the association of the C/EBPß and PU.1 transcription factors and controls Mtb-induced IL-1ß production. The high-IL-1ß expressing genotype was associated with the development of active tuberculosis, the severity of pulmonary disease and poor treatment outcome in TB patients. Higher IL-1ß expression did not suppress the activity of IFN-γ-producing T cells, but instead correlated with neutrophil accumulation in the lung. These observations support a specific role for IL-1ß and granulocytic inflammation as a driver of TB disease progression in humans, and suggest novel strategies for the prevention and treatment of tuberculosis.


Assuntos
Alelos , Proteína beta Intensificadora de Ligação a CCAAT/genética , Suscetibilidade a Doenças/microbiologia , Interleucina-1beta/genética , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Tuberculose/microbiologia , Linhagem Celular , Humanos , Interferon gama/metabolismo , Pulmão/microbiologia
15.
J Immunol ; 190(11): 5722-30, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23630357

RESUMO

Although adjuvants are critical vaccine components, their modes of action are poorly understood. In this study, we investigated the mechanisms by which the heat-killed mycobacteria in CFA promote Th17 CD4(+) T cell responses. We found that IL-17 secretion by CD4(+) T cells following CFA immunization requires MyD88 and IL-1ß/IL-1R signaling. Through measurement of Ag-specific responses after adoptive transfer of OTII cells, we confirmed that MyD88-dependent signaling controls Th17 differentiation rather than simply production of IL-17. Additional experiments showed that CFA-induced Th17 differentiation involves IL-1ß processing by the inflammasome, as mice lacking caspase-1, ASC, or NLRP3 exhibit partially defective responses after immunization. Biochemical fractionation studies further revealed that peptidoglycan is the major component of heat-killed mycobacteria responsible for inflammasome activation. By assaying Il1b transcripts in the injection site skin of CFA-immunized mice, we found that signaling through the adaptor molecule caspase activation and recruitment domain 9 (CARD9) plays a major role in triggering pro-IL-1ß expression. Moreover, we demonstrated that recognition of the mycobacterial glycolipid trehalose dimycolate (cord factor) by the C-type lectin receptor mincle partially explains this CARD9 requirement. Importantly, purified peptidoglycan and cord factor administered in mineral oil synergized to recapitulate the Th17-promoting activity of CFA, and, as expected, this response was diminished in caspase-1- and CARD9-deficient mice. Taken together, these findings suggest a general strategy for the rational design of Th17-skewing adjuvants by combining agonists of the CARD9 pathway with inflammasome activators.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores Corda/imunologia , Lectinas Tipo C/metabolismo , Proteínas de Membrana/metabolismo , Mycobacterium/imunologia , Peptidoglicano/imunologia , Células Th17/imunologia , Células Th17/metabolismo , Adjuvantes Imunológicos , Animais , Proteínas Adaptadoras de Sinalização CARD , Diferenciação Celular/imunologia , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Knockout , Mycobacterium/química , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-18/metabolismo , Transdução de Sinais , Células Th17/citologia , Receptores Toll-Like/metabolismo
16.
Nat Rev Immunol ; 4(2): 100-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15040583

RESUMO

Activation of the innate immune system by interferon-gamma (IFN-gamma is crucial for host resistance to infection. IFN-gamma induces the expression of a wide range of mediators that undermine the ability of pathogens to survive in host cells, including a newly discovered family of 47-kDa GTPases. Elimination of different p47 GTPases in mice by gene targeting severely cripples IFN-gamma-regulated defence against Toxoplasma gondii, Listeria monocytogenes, Mycobacterium spp. and other pathogens. In this article, we review our understanding of the role of p47 GTPases in resistance to intracellular infection and discuss the present evidence concerning their mode of action.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Interferon gama/metabolismo , Viroses/metabolismo , Animais , Infecções Bacterianas/imunologia , GTP Fosfo-Hidrolases/imunologia , Humanos , Imunidade Inata/imunologia , Interferon gama/imunologia , Infecções por Protozoários/imunologia , Viroses/imunologia
17.
iScience ; 27(5): 109807, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38766355

RESUMO

Type I interferon (IFN) production is crucial in tuberculosis pathogenesis, yet the bacterial factors initiating this process are incompletely understood. CpsA, protein of Mycobacterium marinum and Mycobacterium tuberculosis, plays a key role in maintaining bacterial virulence and inhibiting host cell LC3-associated phagocytosis. By utilizing CpsA full deletion mutant studies, we re-verified its essential role in infection-induced pathology and revealed its new role in type I IFN expression. CpsA deficiency hindered IFN production in infected macrophages in vitro as well as zebrafish and mice in vivo. This effect was linked to the cGAS-TBK1-IRF3 pathway, as evidenced by decreased TBK1 and IRF3 phosphorylation in CpsA-deficient bacterial strain-infected macrophages. Moreover, we further show that CpsA deficiency cause decreased cytosolic DNA levels, correlating with impaired phagosomal membrane rupture. Our findings reveal a new function of mycobacterial CpsA in type I IFN production and offer insight into the molecular mechanisms underlying mycobacterial infection pathology.

18.
Nat Microbiol ; 9(7): 1856-1872, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38806671

RESUMO

Adaptation to hypoxia is a major challenge for the survival of Mycobacterium tuberculosis (Mtb) in vivo. Interferon (IFN)-γ-producing CD8+ T cells contribute to control of Mtb infection, in part by promoting antimicrobial activities of macrophages. Whether Mtb counters these responses, particularly during hypoxic conditions, remains unknown. Using metabolomic, proteomic and genetic approaches, here we show that Mtb induced Rv0884c (SerC), an Mtb phosphoserine aminotransferase, to produce D-serine. This activity increased Mtb pathogenesis in mice but did not directly affect intramacrophage Mtb survival. Instead, D-serine inhibited IFN-γ production by CD8+ T cells, which indirectly reduced the ability of macrophages to restrict Mtb upon co-culture. Mechanistically, D-serine interacted with WDR24 and inhibited mTORC1 activation in CD8+ T cells. This decreased T-bet expression and reduced IFN-γ production by CD8+ T cells. Our findings suggest an Mtb evasion mechanism where pathogen metabolic adaptation to hypoxia leads to amino acid-dependent suppression of adaptive anti-TB immunity.


Assuntos
Linfócitos T CD8-Positivos , Interferon gama , Macrófagos , Mycobacterium tuberculosis , Serina , Tuberculose , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Mycobacterium tuberculosis/imunologia , Camundongos , Serina/metabolismo , Interferon gama/metabolismo , Interferon gama/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Tuberculose/imunologia , Tuberculose/microbiologia , Camundongos Endogâmicos C57BL , Transaminases/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Hipóxia/imunologia , Hipóxia/metabolismo , Feminino , Interações Hospedeiro-Patógeno/imunologia
19.
J Exp Med ; 204(2): 273-83, 2007 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-17283209

RESUMO

Although interferon gamma (IFN-gamma) secretion is essential for control of most intracellular pathogens, host survival often also depends on the expression of interleukin 10 (IL-10), a cytokine known to counteract IFN-gamma effector functions. We analyzed the source of regulatory IL-10 in mice infected with the protozoan parasite Toxoplasma gondii. Unexpectedly, IFN-gamma-secreting T-bet(+)Foxp3(-) T helper type 1 (Th1) cells were found to be the major producers of IL-10 in these animals. Further analysis revealed that the same IL-10(+)IFN-gamma(gamma) population displayed potent effector function against the parasite while, paradoxically, also inducing profound suppression of IL-12 production by antigen-presenting cells. Although at any given time point only a fraction of the cells appeared to simultaneously produce IL-10 and IFN-gamma, IL-10 production could be stimulated in IL-10(-)IFN-gamma(+) cells by further activation in vitro. In addition, experiments with T. gondii-specific IL-10(+)IFN-gamma(+) CD4 clones revealed that although IFN-gamma expression is imprinted and triggered with similar kinetics regardless of the state of Th1 cell activation, IL-10 secretion is induced more rapidly from recently activated than from resting cells. These findings indicate that IL-10 production by CD4(+) T lymphocytes need not involve a distinct regulatory Th cell subset but can be generated in Th1 cells as part of the effector response to intracellular pathogens.


Assuntos
Interleucina-10/imunologia , Transdução de Sinais/imunologia , Células Th1/metabolismo , Toxoplasma/imunologia , Toxoplasmose/imunologia , Animais , Anticorpos Monoclonais/imunologia , Citocinas/sangue , Interferon gama/metabolismo , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
20.
J Immunol ; 186(3): 1598-607, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21172867

RESUMO

Although CD4 T cells are required for host resistance to Mycobacterium tuberculosis, they may also contribute to pathology. In this study, we examine the role of the inhibitory receptor PD-1 and its ligand PD-L1 during M. tuberculosis infection. After aerosol exposure, PD-1 knockout (KO) mice develop high numbers of M. tuberculosis-specific CD4 T cells but display markedly increased susceptibility to infection. Importantly, we show that CD4 T cells themselves drive the increased bacterial loads and pathology seen in infected PD-1 KO mice, and PD-1 deficiency in CD4 T cells is sufficient to trigger early mortality. PD-L1 KO mice also display enhanced albeit less severe susceptibility, indicating that T cells are regulated by multiple PD ligands during M. tuberculosis infection. M. tuberculosis-specific CD8 T cell responses were normal in PD-1 KO mice, and CD8 T cells only had a minor contribution to the exacerbated disease in the M. tuberculosis-infected PD-1 KO and PD-L1 KO mice. Thus, in the absence of the PD-1 pathway, M. tuberculosis benefits from CD4 T cell responses, and host resistance requires inhibition by PD-1 to prevent T cell-driven exacerbation of the infection.


Assuntos
Antígenos de Superfície/fisiologia , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/patologia , Aerossóis , Animais , Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Antígeno B7-1/metabolismo , Antígeno B7-1/fisiologia , Antígeno B7-H1 , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Predisposição Genética para Doença , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Necrose , Peptídeos/metabolismo , Peptídeos/fisiologia , Receptor de Morte Celular Programada 1 , Tuberculose Pulmonar/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA