Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nature ; 474(7351): 385-9, 2011 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-21602824

RESUMO

Live vaccines have long been known to trigger far more vigorous immune responses than their killed counterparts. This has been attributed to the ability of live microorganisms to replicate and express specialized virulence factors that facilitate invasion and infection of their hosts. However, protective immunization can often be achieved with a single injection of live, but not dead, attenuated microorganisms stripped of their virulence factors. Pathogen-associated molecular patterns (PAMPs), which are detected by the immune system, are present in both live and killed vaccines, indicating that certain poorly characterized aspects of live microorganisms, not incorporated in dead vaccines, are particularly effective at inducing protective immunity. Here we show that the mammalian innate immune system can directly sense microbial viability through detection of a special class of viability-associated PAMPs (vita-PAMPs). We identify prokaryotic messenger RNA as a vita-PAMP present only in viable bacteria, the recognition of which elicits a unique innate response and a robust adaptive antibody response. Notably, the innate response evoked by viability and prokaryotic mRNA was thus far considered to be reserved for pathogenic bacteria, but we show that even non-pathogenic bacteria in sterile tissues can trigger similar responses, provided that they are alive. Thus, the immune system actively gauges the infectious risk by searching PAMPs for signatures of microbial life and thus infectivity. Detection of vita-PAMPs triggers a state of alert not warranted for dead bacteria. Vaccine formulations that incorporate vita-PAMPs could thus combine the superior protection of live vaccines with the safety of dead vaccines.


Assuntos
Imunidade Inata/imunologia , Viabilidade Microbiana/genética , Viabilidade Microbiana/imunologia , RNA Bacteriano/imunologia , RNA Mensageiro/imunologia , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Bactérias/genética , Bactérias/imunologia , Bactérias/patogenicidade , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Proteínas de Transporte/metabolismo , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interferon beta/genética , Interferon beta/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fagocitose , Fagossomos/imunologia , Fagossomos/microbiologia , RNA Bacteriano/genética , RNA Mensageiro/genética , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas de Produtos Inativados/imunologia , Fatores de Virulência
2.
Mol Med ; 21(1): 1025-1037, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26623925

RESUMO

Although the molecular links underlying the causative relationship between chronic low-grade inflammation and insulin resistance are not completely understood, compelling evidence suggests a pivotal role of the nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Here we tested the hypothesis that either a selective pharmacological inhibition or a genetic downregulation of the NLRP3 inflammasome results in reduction of the diet-induced metabolic alterations. Male C57/BL6 wild-type mice and NLRP3-/- littermates were fed control diet or high-fat, high-fructose diet (HD). A subgroup of HD-fed wild-type mice was treated with the NLRP3 inflammasome inhibitor BAY 11-7082 (3 mg/kg intraperitoneally [IP]). HD feeding increased plasma and hepatic lipids and impaired glucose homeostasis and renal function. Renal and hepatic injury was associated with robust increases in profibrogenic markers, while only minimal fibrosis was recorded. None of these metabolic abnormalities were detected in HD-fed NLRP3-/- mice, and they were dramatically reduced in HD-mice treated with the NLRP3 inflammasome inhibitor. BAY 11-7082 also attenuated the diet-induced increase in NLRP3 inflammasome expression, resulting in inhibition of caspase-1 activation and interleukin (IL)-1ß and IL-18 production (in liver and kidney). Interestingly, BAY 11-7082, but not gene silencing, inhibited nuclear factor (NF)-κB nuclear translocation. Overall, these results demonstrate that the selective pharmacological modulation of the NLRP3 inflammasome attenuates the metabolic abnormalities and the related organ injury/dysfunction caused by chronic exposure to HD, with effects similar to those obtained by NLRP3 gene silencing.

3.
J Immunol ; 193(4): 1787-98, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25024388

RESUMO

Deciphering the mechanisms that allow the induction of strong immune responses is crucial to developing efficient vaccines against infectious diseases and cancer. Based on the discovery that the adenylate cyclase from Bordetella pertussis binds to the CD11b/CD18 integrin, we developed a highly efficient detoxified adenylate cyclase-based vector (CyaA) capable of delivering a large variety of Ags to the APC. This vector allows the induction of protective and therapeutic immunity against viral and tumoral challenges as well as against transplanted tumors in the absence of any added adjuvant. Two therapeutic vaccine candidates against human papilloma viruses and melanoma have been developed recently, based on the CyaA vector, and are currently in clinical trials. We took advantage of one of these highly purified vaccines, produced under good manufacturing practice-like conditions, to decipher the mechanisms by which CyaA induces immune responses. In this study, we demonstrate that CyaA binds both human and mouse CD11b(+) dendritic cells (DCs) and induces their maturation, as shown by the upregulation of costimulatory and MHC molecules and the production of proinflammatory cytokines. Importantly, we show that DCs sense CyaA through the TLR4/Toll/IL-1R domain-containing adapter-inducing IFN-ß pathway, independent of the presence of LPS. These findings show that CyaA possesses the intrinsic ability to not only target DCs but also to activate them, leading to the induction of strong immune responses. Overall, this study demonstrates that Ag delivery to CD11b(+) DCs in association with TLR4/Toll/IL-1R domain-containing adapter-inducing IFN-ß activation is an efficient strategy to promote strong specific CD8(+) T cell responses.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Toxina Adenilato Ciclase/imunologia , Antígeno CD11b/imunologia , Células Dendríticas/imunologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Bordetella pertussis/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/citologia , Feminino , Interferon beta/imunologia , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptores de Interleucina-1/imunologia , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/imunologia , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Tirosina/genética
4.
Infect Immun ; 83(9): 3612-23, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26123801

RESUMO

Tumor necrosis factor (TNF) is an important cytokine for host defense against pathogens but is also associated with the development of human immunopathologies. TNF blockade effectively ameliorates many chronic inflammatory conditions but compromises host immunity to tuberculosis. The search for novel, more specific human TNF blockers requires the development of a reliable animal model. We used a novel mouse model with complete replacement of the mouse TNF gene by its human ortholog (human TNF [huTNF] knock-in [KI] mice) to determine resistance to Mycobacterium bovis BCG and M. tuberculosis infections and to investigate whether TNF inhibitors in clinical use reduce host immunity. Our results show that macrophages from huTNF KI mice responded to BCG and lipopolysaccharide similarly to wild-type macrophages by NF-κB activation and cytokine production. While TNF-deficient mice rapidly succumbed to mycobacterial infection, huTNF KI mice survived, controlling the bacterial burden and activating bactericidal mechanisms. Administration of TNF-neutralizing biologics disrupted the control of mycobacterial infection in huTNF KI mice, leading to an increased bacterial burden and hyperinflammation. Thus, our findings demonstrate that human TNF can functionally replace murine TNF in vivo, providing mycobacterial resistance that could be compromised by TNF neutralization. This new animal model will be helpful for the testing of specific biologics neutralizing human TNF.


Assuntos
Modelos Animais de Doenças , Infecções por Mycobacterium/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Animais , Western Blotting , Citocinas/biossíntese , Citometria de Fluxo , Técnicas de Introdução de Genes/métodos , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium bovis/imunologia , Mycobacterium tuberculosis/imunologia
5.
Nat Med ; 13(9): 1050-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17704786

RESUMO

Conventional cancer treatments rely on radiotherapy and chemotherapy. Such treatments supposedly mediate their effects via the direct elimination of tumor cells. Here we show that the success of some protocols for anticancer therapy depends on innate and adaptive antitumor immune responses. We describe in both mice and humans a previously unrecognized pathway for the activation of tumor antigen-specific T-cell immunity that involves secretion of the high-mobility-group box 1 (HMGB1) alarmin protein by dying tumor cells and the action of HMGB1 on Toll-like receptor 4 (TLR4) expressed by dendritic cells (DCs). During chemotherapy or radiotherapy, DCs require signaling through TLR4 and its adaptor MyD88 for efficient processing and cross-presentation of antigen from dying tumor cells. Patients with breast cancer who carry a TLR4 loss-of-function allele relapse more quickly after radiotherapy and chemotherapy than those carrying the normal TLR4 allele. These results delineate a clinically relevant immunoadjuvant pathway triggered by tumor cell death.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Receptor 4 Toll-Like/imunologia , Animais , Neoplasias Ósseas/tratamento farmacológico , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/radioterapia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Compostos Organoplatínicos/uso terapêutico , Osteossarcoma/tratamento farmacológico , Piridinas/uso terapêutico
6.
Part Fibre Toxicol ; 11: 69, 2014 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-25497724

RESUMO

BACKGROUND: Inflammasome-activated IL-1ß plays a major role in lung neutrophilic inflammation induced by inhaled silica. However, the exact mechanisms that contribute to the initial production of precursor IL-1ß (pro-IL-1ß) are still unclear. Here, we assessed the implication of alarmins (IL-1α, IL-33 and HMGB1) in the lung response to silica particles and found that IL-1α is a master cytokine that regulates IL-1ß expression. METHODS: Pro- and mature IL-1ß as well as alarmins were assessed by ELISA, Western Blot or qRT-PCR in macrophage cultures and in mouse lung following nano- and micrometric silica exposure. Implication of these immune mediators in the establishment of lung inflammatory responses to silica was investigated in knock-out mice or after antibody blockade by evaluating pulmonary neutrophil counts, CXCR2 expression and degree of histological injury. RESULTS: We found that the early release of IL-1α and IL-33, but not HMGB1 in alveolar space preceded the lung expression of pro-IL-1ß and neutrophilic inflammation in silica-treated mice. In vitro, the production of pro-IL-1ß by alveolar macrophages was significantly induced by recombinant IL-1α but not by IL-33. Neutralization or deletion of IL-1α reduced IL-1ß production and neutrophil accumulation after silica in mice. Finally, IL-1α released by J774 macrophages after in vitro exposure to a range of micro- and nanoparticles of silica was correlated with the degree of lung inflammation induced in vivo by these particles. CONCLUSIONS: We demonstrated that in response to silica exposure, IL-1α is rapidly released from pre-existing stocks in alveolar macrophages and promotes subsequent lung inflammation through the stimulation of IL-1ß production. Moreover, we demonstrated that in vitro IL-1α release from macrophages can be used to predict the acute inflammogenic activity of silica micro- and nanoparticles.


Assuntos
Poluentes Atmosféricos/toxicidade , Exposição por Inalação/efeitos adversos , Interleucina-1alfa/metabolismo , Pulmão/efeitos dos fármacos , Nanopartículas/toxicidade , Pneumonia/induzido quimicamente , Dióxido de Silício/toxicidade , Poluentes Atmosféricos/química , Animais , Anticorpos Neutralizantes/metabolismo , Linhagem Celular , Células Cultivadas , Feminino , Interleucina-1alfa/antagonistas & inibidores , Interleucina-1alfa/genética , Interleucina-1beta/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Macrófagos Alveolares/citologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microesferas , Nanopartículas/administração & dosagem , Nanopartículas/química , Infiltração de Neutrófilos/efeitos dos fármacos , Tamanho da Partícula , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Dióxido de Silício/administração & dosagem , Dióxido de Silício/química , Testes de Toxicidade Aguda
7.
Nat Med ; 12(2): 214-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16444265

RESUMO

The interferon (IFN)-gamma-induced TRAIL effector mechanism is a vital component of cancer immunosurveillance by natural killer (NK) cells in mice. Here we show that the main source of IFN-gamma is not the conventional NK cell but a subset of B220(+)Ly6C(-) dendritic cells, which are atypical insofar as they express NK cell-surface molecules. Upon contact with a variety of tumor cells that are poorly recognized by NK cells, B220(+)NK1.1(+) dendritic cells secrete high levels of IFN-gamma and mediate TRAIL-dependent lysis of tumor cells. Adoptive transfer of these IFN-producing killer dendritic cells (IKDCs) into tumor-bearing Rag2(-/-)Il2rg(-/-) mice prevented tumor outgrowth, whereas transfer of conventional NK cells did not. In conclusion, we identified IKDCs as pivotal sensors and effectors of the innate antitumor immune response.


Assuntos
Células Dendríticas/classificação , Células Dendríticas/imunologia , Neoplasias Experimentais/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno , Antígenos Ly , Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/imunologia , Antígeno CD11c/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Células Dendríticas/ultraestrutura , Feminino , Interferon gama/biossíntese , Subunidade gama Comum de Receptores de Interleucina , Células Matadoras Naturais/imunologia , Lectinas Tipo C/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Microscopia Eletrônica , Subfamília B de Receptores Semelhantes a Lectina de Células NK , Receptores de Interleucina/deficiência , Receptores de Interleucina/genética , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/imunologia
8.
Mol Ther ; 20(8): 1571-81, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22643865

RESUMO

The administration of recombinant adeno-associated viral vectors (rAAV) for gene transfer induces strong humoral responses through mechanisms that remain incompletely characterized. To investigate the links between innate and adaptive immune responses to the vector, rAAVs were injected intravenously into mice deficient in cell-intrinsic components of innate responses (Toll-like receptors (TLRs), type-1 interferon (IFN) or inflammasome signaling molecules) and AAV-specific antibodies were measured. Of all molecules tested, only MyD88 was critically needed to mount immunoglobulin G (IgG) responses since MyD88(-/-) mice failed to develop high levels of AAV-specific IgG2 and IgG3, regardless of capsid serotype injected. None of the TLRs tested was essential here, but TLR9 ensured a Th1-biased antibody responses. Indeed, capsid-specific Th1 cells were induced upon injection of rAAV1, as directly confirmed with an epitope-tagged capsid, and the priming and development of these Th1 cells required T cell-extrinsic MyD88. Cell transfer experiments showed that autonomous MyD88 signaling in B cells, but not T cells, was sufficient to produce Th1-dependent IgGs. Therefore, rAAV triggers innate responses, at least via B cells, controlling the development of capsid-specific Th1-driven antibodies. MyD88 emerges as a critical and pivotal regulator of both T- and B-cell adaptive immunity against AAV.


Assuntos
Adenoviridae/imunologia , Anticorpos/imunologia , Anticorpos/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Células Th1/imunologia , Animais , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
9.
J Infect Dis ; 206(5): 723-34, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22723642

RESUMO

BACKGROUND: Exogenous activation of pulmonary invariant natural killer T (iNKT) cells, a population of lipid-reactive αß T lymphocytes, with use of mucosal α-galactosylceramide (α-GalCer) administration, is a promising approach to control respiratory bacterial infections. We undertook the present study to characterize mechanisms leading to α-GalCer-mediated protection against lethal infection with Streptococcus pneumoniae serotype 1, a major respiratory pathogen in humans. METHODS AND RESULTS: α-GalCer was administered by the intranasal route before infection with S. pneumoniae. We showed that respiratory dendritic cells (DCs), most likely the CD103(+) subset, play a major role in the activation (IFN-γ and IL-17 release) of pulmonary iNKT cells, whereas alveolar and interstitial macrophages are minor players. After challenge, S. pneumoniae was rapidly (4 hours) eliminated in the alveolar spaces, a phenomenon that depended on respiratory DCs and neutrophils, but not macrophages, and on the early production of both IFN-γ and IL-17. Protection was also associated with the synthesis of various interferon-dependent and IL-17-associated genes as revealed by transcriptomic analysis. CONCLUSIONS: These data imply a new function for pulmonary CD103(+) DCs in mucosal activation of iNKT cells and establish a critical role for both IFN-γ and IL-17 signalling pathways in mediating the innate immune response to S. pneumoniae.


Assuntos
Células Dendríticas/imunologia , Galactosilceramidas/farmacologia , Células T Matadoras Naturais/imunologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Animais , Antígenos CD/imunologia , Líquido da Lavagem Broncoalveolar/microbiologia , Células Dendríticas/microbiologia , Galactosilceramidas/uso terapêutico , Imunidade Inata/imunologia , Cadeias alfa de Integrinas/imunologia , Interferon gama/imunologia , Interleucina-17/imunologia , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células T Matadoras Naturais/microbiologia , Infecções Pneumocócicas/microbiologia , Transdução de Sinais
10.
J Immunol ; 184(11): 6367-77, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20421647

RESUMO

IL-17-producing T lymphocytes play a crucial role in inflammation, but their possible implication in fibrosis remains to be explored. In this study, we examined the involvement of these cells in a mouse model of lung inflammation and fibrosis induced by silica particles. Upregulation of IL-17A was associated with the development of experimental silicosis, but this response was markedly reduced in athymic, gammadelta T cell-deficient or CD4(+) T cell-depleted mice. In addition, gammadelta T lymphocytes and CD4(+) T cells, but not macrophages, neutrophils, NK cells or CD8 T cells, purified from the lungs of silicotic mice markedly expressed IL-17A. Depletion of alveolar macrophages or neutralization of IL-23 reduced upregulation of IL-17A in the lung of silicotic mice. IL-17R-deficient animals (IL-17R(-/-)) or IL-17A Ab neutralization, but not IL-22(-/-) mice, developed reduced neutrophil influx and injury during the early lung response to silica. However, chronic inflammation, fibrosis, and TGF-beta expression induced by silica were not attenuated in the absence of IL-17R or -22 or after IL-17A Ab blockade. In conclusion, a rapid lung recruitment of IL-17A-producing T cells, mediated by macrophage-derived IL-23, is associated with experimental silicosis in mice. Although the acute alveolitis induced by silica is IL-17A dependent, this cytokine appears dispensable for the development of the late inflammatory and fibrotic lung responses to silica.


Assuntos
Interleucina-17/imunologia , Pneumonia/imunologia , Fibrose Pulmonar/imunologia , Silicose/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Separação Celular , Modelos Animais de Doenças , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Pneumonia/patologia , Reação em Cadeia da Polimerase , Fibrose Pulmonar/patologia , RNA Mensageiro/análise , Receptores de Antígenos de Linfócitos T gama-delta , Silicose/patologia
11.
Microbiology (Reading) ; 157(Pt 3): 774-785, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21071495

RESUMO

Finding two or more genotypes of a single species within an infected sample is a not infrequent event. In this work, three Escherichia coli strains of decreasing extra-intestinal virulence (pathogenic B2S and B1S strains, and the avirulent K-12 MG1655 strain) were tested in septicaemia and urinary tract infection (UTI) mouse models, either separately or in pairs. Survival was monitored and bacteria were counted in various organs. Serum interleukin (IL)-6, tumour necrosis factor alpha (TNFα) and IL-10 were measured. We show that a mix of high amounts of B1S or of MG1655 with low amounts of B2S killed more rapidly (B1S), or killed more mice (MG1655), than either high amounts of B1S, high amounts of MG1655 or low amounts of B2S separately in the mouse septicaemia model. This bacterial synergy persisted when high amounts of dead or abnormal-LPS K-12 cells were injected together with a low amount of B2S. In both septicaemia and UTI models, significantly more bacteria were recovered from the organs of mice injected with the MG1655/B2S mix than from those of mice injected with the inocula separately. Consistently, in the septicaemia model, more IL-6 was secreted before death by the mice that were injected with the mix of bacteria than by the mice that were injected with the inocula separately. The synergistically enhanced mortality in the case of co-infection in the septicaemia model persisted in RFcγ(-/-), Myd88(-/-) and IL-6(-/-) knockout mice. This synergistically increased virulence resulting from the interaction between an avirulent and a pathogenic strain of the same bacterial species raises questions about the role of avirulent bacteria in the development of some extra-intestinal infections.


Assuntos
Bacteriemia/mortalidade , Infecções por Escherichia coli/mortalidade , Escherichia coli K12/fisiologia , Escherichia coli/patogenicidade , Infecções Urinárias/mortalidade , Animais , Bacteriemia/microbiologia , Bacteriemia/patologia , Modelos Animais de Doenças , Escherichia coli/classificação , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Escherichia coli K12/genética , Escherichia coli K12/isolamento & purificação , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Infecções Urinárias/microbiologia , Infecções Urinárias/patologia , Virulência
12.
PLoS Pathog ; 4(5): e1000078, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18516300

RESUMO

African trypanosomes of the Trypanosoma brucei species are extra-cellular parasites that cause human African trypanosomiasis (HAT) as well as infections in game animals and livestock. Trypanosomes are known to evade the immune response of their mammalian host by continuous antigenic variation of their surface coat. Here, we aim to demonstrate that in addition, trypanosomes (i) cause the loss of various B cell populations, (ii) disable the hosts' capacity to raise a long-lasting specific protective anti-parasite antibody response, and (iii) abrogate vaccine-induced protective response to a non-related human pathogen such as Bordetella pertussis. Using a mouse model for T. brucei, various B cell populations were analyzed by FACS at different time points of infection. The results show that during early onset of a T. brucei infection, spleen remodeling results in the rapid loss of the IgM(+) marginal zone (IgM(+)MZ) B cell population characterized as B220(+)IgM(High)IgD(Int) CD21(High)CD23(Low)CD1d(+)CD138(-). These cells, when isolated during the first peak of infection, stained positive for Annexin V and had increased caspase-3 enzyme activity. Elevated caspase-3 mRNA levels coincided with decreased mRNA levels of the anti-apoptotic Bcl-2 protein and BAFF receptor (BAFF-R), indicating the onset of apoptosis. Moreover, affected B cells became unresponsive to stimulation by BCR cross-linking with anti-IgM Fab fragments. In vivo, infection-induced loss of IgM(+) B cells coincided with the disappearance of protective variant-specific T-independent IgM responses, rendering the host rapidly susceptible to re-challenge with previously encountered parasites. Finally, using the well-established human diphtheria, tetanus, and B. pertussis (DTPa) vaccination model in mice, we show that T. brucei infections abrogate vaccine-induced protective responses to a non-related pathogen such as B. pertussis. Infections with T. brucei parasites result in the rapid loss of T-cell independent IgM(+)MZ B cells that are normally functioning as the primary immune barrier against blood-borne pathogens. In addition, ongoing trypanosome infections results in the rapid loss of B cell responsiveness and prevent the induction of protective memory responses. Finally, trypanosome infections disable the host's capacity to recall vaccine-induced memory responses against non-related pathogens. In particular, these last results call for detailed studies of the effect of HAT on memory recall responses in humans, prior to the planning of any mass vaccination campaign in HAT endemic areas.


Assuntos
Apoptose , Linfócitos B/patologia , Interações Hospedeiro-Parasita , Memória Imunológica/imunologia , Tripanossomíase Africana/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Antiprotozoários/biossíntese , Proliferação de Células , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Modelos Animais de Doenças , Citometria de Fluxo , Expressão Gênica , Imunofenotipagem , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/metabolismo , Baço/imunologia , Baço/patologia , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei brucei/patogenicidade , Vacinas/administração & dosagem
13.
Nat Med ; 26(6): 919-931, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32451498

RESUMO

The prognosis of colon cancer (CC) is dictated by tumor-infiltrating lymphocytes, including follicular helper T (TFH) cells and the efficacy of chemotherapy-induced immune responses. It remains unclear whether gut microbes contribute to the elicitation of TFH cell-driven responses. Here, we show that the ileal microbiota dictates tolerogenic versus immunogenic cell death of ileal intestinal epithelial cells (IECs) and the accumulation of TFH cells in patients with CC and mice. Suppression of IEC apoptosis led to compromised chemotherapy-induced immunosurveillance against CC in mice. Protective immune responses against CC were associated with residence of Bacteroides fragilis and Erysipelotrichaceae in the ileum. In the presence of these commensals, apoptotic ileal IECs elicited PD-1+ TFH cells in an interleukin-1R1- and interleukin-12-dependent manner. The ileal microbiome governed the efficacy of chemotherapy and PD-1 blockade in CC independently of microsatellite instability. These findings demonstrate that immunogenic ileal apoptosis contributes to the prognosis of chemotherapy-treated CC.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Microbioma Gastrointestinal/imunologia , Íleo/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Oxaliplatina/farmacologia , Adenocarcinoma/imunologia , Adenocarcinoma/microbiologia , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos/uso terapêutico , Apoptose/imunologia , Bacteroides fragilis , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/microbiologia , Neoplasias do Colo/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Firmicutes , Microbioma Gastrointestinal/fisiologia , Humanos , Íleo/imunologia , Íleo/microbiologia , Íleo/patologia , Morte Celular Imunogênica/efeitos dos fármacos , Morte Celular Imunogênica/imunologia , Vigilância Imunológica/efeitos dos fármacos , Vigilância Imunológica/imunologia , Interleucina-12/imunologia , Mucosa Intestinal , Linfócitos do Interstício Tumoral/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Oxaliplatina/uso terapêutico , Prognóstico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptores Tipo I de Interleucina-1/imunologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia
14.
Arthritis Res Ther ; 20(1): 119, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29884199

RESUMO

BACKGROUND: Epidemiologic studies have highlighted the association of environmental factors with the development and progression of autoimmune and chronic inflammatory diseases. Among the environmental factors, smoking has been associated with increased susceptibility and poor prognosis in rheumatoid arthritis (RA). However, the immune and molecular mechanism of smoking-induced arthritis aggravation remains unclear. The transcription factor aryl hydrocarbon receptor (AHR) regulates the generation of Th17 cells, CD4 T cells linked the development of autoimmune diseases. AHR is activated by organic compounds including polycyclic aromatic hydrocarbons (PAHs), which are environmental pollutants that are also present in cigarette smoke. In this study, we investigated the role of AHR activation in the aggravation of experiment arthritis induced by exposure to cigarette smoke. METHODS: Mice were exposed to cigarette smoke during the developmental phase of antigen-induced arthritis and collagen-induced arthritis to evaluate the effects of smoking on disease development. Aggravation of articular inflammation was assessed by measuring neutrophil migration to the joints, increase in articular hyperalgesia and changes in the frequencies of Th17 cells. In vitro studies were performed to evaluate the direct effects of cigarette smoke and PAH on Th17 differentiation. We also used mice genetically deficient for AHR (Ahr KO) and IL-17Ra (Il17ra KO) to determine the in vivo mechanism of smoking-induced arthritis aggravation. RESULTS: We found that smoking induces arthritis aggravation and increase in the frequencies of Th17 cells. The absence of IL-17 signaling (Il17ra KO) conferred protection to smoking-induced arthritis aggravation. Moreover, in vitro experiments showed that cigarette smoke can directly increase Th17 differentiation of T cells by inducing AHR activation. Indeed, Ahr KO mice were protected from cigarette smoke-induced arthritis aggravation and did not display increase in TH17 frequencies, suggesting that AHR activation is an important mechanism for cigarette smoke effects on arthritis. Finally, we demonstrate that PAHs are also able to induce arthritis aggravation. CONCLUSIONS: Our data demonstrate that the disease-exacerbating effects of cigarette smoking are AHR dependent and environmental pollutants with AHR agonist activity can induce arthritis aggravation by directly enhancing Th17 cell development.


Assuntos
Artrite Experimental/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Fumaça/efeitos adversos , Células Th17/metabolismo , Animais , Artrite Experimental/etiologia , Artrite Experimental/genética , Compostos Azo/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Pirazóis/farmacologia , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Receptores de Hidrocarboneto Arílico/genética , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Nicotiana/química
15.
Immun Inflamm Dis ; 5(2): 200-213, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28474504

RESUMO

INTRODUCTION: Viruses have developed multiple mechanisms to alter immune reactions. In 1969, it was reported that lactate dehydrogenase-elevating virus (LDV), a single stranded positive sense mouse nidovirus, delays skin allograft rejection and inhibits spleen alterations in graft versus host disease (GVHD). As the underlying mechanisms have remained unresolved and given the need for new therapies of this disease, we reassessed the effects of the virus on GVHD and tried to uncover its mode of action. METHODS: GVHD was induced by transfer of parent (B6) spleen cells to non-infected or LDV-infected B6D2F1 recipients. In vitro mixed-lymhocyte culture (MLC) reactions were used to test the effects of the virus on antigen-presenting cells (APC) and responder T cells. RESULTS: LDV infection resulted in a threefold increase in survival rate with reduced weight loss and liver inflammation but with the establishment of permanent chimerism that correlated with decreased interleukine (IL)-27 and interferon (IFN)γ plasma levels. Infected mice showed a transient elimination of splenic CD11b+ and CD8α+ conventional dendritic cells (cDCs) required for allogeneic CD4 and CD8 T cell responses in vitro. This drop of APC numbers was not observed with APCs derived from toll-like receptor (TLR)7-deficient mice. A second effect of the virus was a decreased T cell proliferation and IFNγ production during MLC without detectable changes in Foxp3+ regulatory T cell (Tregs) numbers. Both cDC and responder T cell inhibition were type I IFN dependent. Although the suppressive effects were very transient, the GVHD inhibition was long-lasting. CONCLUSION: A type I IFN-dependent suppression of DC and T cells just after donor spleen cell transplantation induces permanent chimerism and donor cell implantation in a parent to F1 spleen cell transplantation model. If this procedure can be extended to full allogeneic bone marrow transplantation, it could open new therapeutic perspectives for hematopoietic stem cell transplantation (HSCT).


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/imunologia , Interferon Tipo I/imunologia , Infecções por Nidovirales/imunologia , Nidovirales/imunologia , Aloenxertos , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Células Dendríticas/patologia , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/patologia , Interferon Tipo I/genética , Camundongos , Camundongos Knockout
16.
J Endotoxin Res ; 9(6): 381-4, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14733725

RESUMO

Innate immunity initiates protection of the host organism against invasion of micro-organisms by specific recognition mechanisms. This article reviews the dual role of LBP/CD14 in innate immunity, focusing mostly on experiments performed in mice by the authors. LPS induces uncontrolled pro-inflammatory response that kills the host and is LBP- and CD14-dependent, as neutralization of LBP and CD14 prevents lethal shock. However, surprisingly, the synthetic Pam3CysSerLys4 bacterial lipoprotein from Escherichia coli (BLP), which is well tolerated in mice, kills the mice upon LBP or CD14 blockade. Furthermore, after blockade of LBP and CD14, the mice succumb to a challenge with virulent Klebsiella pneumoniae or Salmonella typhimurium. Therefore, host responses to Gram-negative bacteria are not identical to that of LPS or BLP. When the host is in the presence of virulent Gram-negative bacteria, the invading pathogens must be held in check by the innate immune system until a specific immune response is mounted. Under these conditions, LBP, CD14, and likely Toll-like receptors (TLRs) are a prerequisite to trigger a pro-inflammatory response of macrophages, which is crucial for keeping an infection under control. These studies indicate that we are very far from understanding how the innate system works and more work needs to be done concerning LBP, CD14 or TLRs. Therefore, caution should be the rule about the use of therapeutic approaches to block the pro-inflammatory response in Gram-negative infections.


Assuntos
Endotoxinas/imunologia , Bactérias Gram-Negativas/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/sangue , Lipopolissacarídeos/metabolismo , Animais , Endotoxinas/sangue , Infecções por Bactérias Gram-Negativas/imunologia , Humanos , Imunidade Inata , Inflamação , Camundongos , Camundongos Knockout
17.
PLoS One ; 9(7): e99383, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25050810

RESUMO

The exact implication of innate immunity in granuloma formation and irreversible lung fibrosis remains to be determined. In this study, we examined the lung inflammatory and fibrotic responses to silica in MyD88-knockout (KO) mice. In comparison to wild-type (WT) mice, we found that MyD88-KO animals developed attenuated lung inflammation, neutrophil accumulation and IL-1ß release in response to silica. Granuloma formation was also less pronounced in MyD88-KO mice after silica. This limited inflammatory response was not accompanied by a concomitant attenuation of lung collagen accumulation after silica. Histological analyses revealed that while pulmonary fibrosis was localized in granulomas in WT animals, it was diffusely distributed throughout the parenchyma in MyD88-KO mice. Robust collagen accumulation was also observed in mice KO for several other components of innate immunity (IL-1R, IL-1, ASC, NALP3, IL-18R, IL-33R, TRIF, and TLR2-3-4,). We additionally show that pulmonary fibrosis in MyD88-KO mice was associated with the accumulation of pro-fibrotic regulatory T lymphocytes (T regs) and pro-fibrotic cytokine expression (TGF-ß, IL-10 and PDGF-B), not with T helper (Th) 17 cell influx. Our findings indicate that the activation of MyD88-related innate immunity is central in the establishment of particle-induced lung inflammatory and granuloma responses. The development of lung fibrosis appears uncoupled from inflammation and may be orchestrated by a T reg-associated pathway.


Assuntos
Inflamação/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Fibrose Pulmonar/imunologia , Dióxido de Silício/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Citometria de Fluxo , Granuloma/genética , Granuloma/imunologia , Granuloma/metabolismo , Imunidade Inata/genética , Imunidade Inata/imunologia , Imuno-Histoquímica , Inflamação/induzido quimicamente , Inflamação/genética , Interleucina-1alfa/genética , Interleucina-1alfa/imunologia , Interleucina-1alfa/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Dióxido de Silício/toxicidade , Silicose/etiologia , Silicose/genética , Silicose/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
18.
PLoS One ; 9(2): e87822, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24516564

RESUMO

Conjugated vaccines consisting of flagellin and antigen activate TLR5 and induce strong innate and adaptive immune responses. Objective of the present study was to gain further insight into the mechanisms by which flagellin fusion proteins mediate their immune modulating effects. In a mouse model of Ova-induced intestinal allergy a fusion protein of flagellin and Ova (rflaA:Ova) was used for intranasal and intraperitoneal vaccination. Aggregation status of flaA, Ova and flaA:Ova were compared by light scattering, uptake of fluorescence labeled proteins into mDC was analyzed, processing was investigated by microsomal digestion experiments. Mechanism of DC-activation was investigated using proteasome and inflammasome inhibitors. Immune responses of wildtype, IL-10(-/-), TLR5(-/-) mDCs and Ova-transgenic T cells were investigated. Mucosal and i.p.-application of rflaA:Ova were able to prevent allergic sensitization, suppress disease-related symptoms, prevent body weight loss and reduction in food uptake. Intranasal vaccination resulted in strongest suppression of Ova-specific IgE production. These protective effects were associated with increased aggregation of rflaA:Ova and accompanied by tenfold higher uptake rates into mDC compared to the mixture of both proteins. Microsomal digestion showed that stimulation with rflaA:Ova resulted in faster degradation and the generation of different peptides compared to rOva. rflaA:Ova-mediated activation of mDC could be suppressed in a dose-dependent manner by the application of both inflammasome and proteasome inhibitors. Using TLR5(-/-) mDC the rflaA:Ova induced IL-10 secretion was shown to be TLR5 dependent. In co-cultures of IL-10(-/-) mDC with DO11.10 T cells the lack of rflaA:Ova-mediated IL-10 secretion resulted in enhanced levels of both TH2 (IL-4, IL-5) and TH1 (IL-2 and IFN-y) cytokines. In summary, mucosal vaccination with flaA:Ova showed strongest preventive effect. Stimulation with rflaA:Ova results in strong immune modulation mediated by enhanced uptake of the aggregated fusion protein, likely resulting in a different processing by DC as well as stronger TLR5 mediated cell activation.


Assuntos
Apresentação de Antígeno/imunologia , Hipersensibilidade/prevenção & controle , Interleucina-10/imunologia , Intestinos/imunologia , Receptor 5 Toll-Like/imunologia , Vacinação/métodos , Imunidade Adaptativa/imunologia , Animais , Modelos Animais de Doenças , Camundongos
19.
PLoS One ; 8(3): e58452, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23505509

RESUMO

IL-17A induces the release of pro-inflammatory cytokines and of reactive oxygen species which could lead to neutrophilic inflammation. We determined the role of IL-17 receptor (IL-17R) signalling in oxidant-induced lung emphysema and airway hyperresponsiveness. IL-17R(-/-) and wild-type C57/BL6 mice were exposed to ozone (3 ppm; 3 hours) for 12 times over 6 weeks. Bronchial responsiveness to acetylcholine was measured, and lungs were retrieved. Mean linear intercept (Lm) and isometric contractile responses of intrapulmonary airways to acetylcholine were determined. In wild-type mice but not in IL-17R(-/-), chronic ozone exposure caused airway hyperresponsiveness. The increase in Lm after chronic ozone exposure of wild-type mice was also observed in IL-17R(-/-) mice. The increased maximal contractile response to acetylcholine seen in airways of wild-type mice exposed to ozone was abolished in IL-17R(-/-) mice. p38-mitogen-activated protein kinase (MAPK) and dexamethasone-dependent increase in contractile response was reduced in airways from IL-17R(-/-) ozone-exposed mice. Lung inflammation scores were not altered in IL-17R(-/-) mice exposed to ozone compared to wild-type mice. The increased release of IL-17 and IL-1ß, and the activation of p38 MAPK in the lungs of ozone-exposed mice was reduced in IL-17R(-/-) mice. IL-17R signalling underlies the increase in airway hyperresponsiveness seen after ozone exposure, mediated by the increased contractility of airway smooth muscle. The emphysema and lung inflammation induced by ozone is not dependent on IL-17.


Assuntos
Hiper-Reatividade Brônquica/metabolismo , Enfisema/metabolismo , Interleucina-17/metabolismo , Estresse Oxidativo , Acetilcolina/farmacologia , Resistência das Vias Respiratórias/efeitos dos fármacos , Animais , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Hiper-Reatividade Brônquica/genética , Citocinas/metabolismo , Dexametasona/farmacologia , Enfisema/genética , Inflamação/genética , Inflamação/metabolismo , Contração Isométrica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
20.
Cancer Res ; 72(11): 2757-67, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22427351

RESUMO

During cancer development, a number of regulatory cell subsets and immunosuppressive cytokines subvert adaptive immune responses. Although it has been shown that tumor-derived interleukin (IL)-18 participates in the PD-1-dependent tumor progression in NK cell-controlled cancers, the mechanistic cues underlying this immunosuppression remain unknown. Here, we show that IL-18 converts a subset of Kit(-) (CD11b(-)) into Kit(+) natural killer (NK) cells, which accumulate in all lymphoid organs of tumor bearers and mediate immunoablative functions. Kit(+) NK cells overexpressed B7-H1/PD-L1, a ligand for PD-1. The adoptive transfer of Kit(+) NK cells promoted tumor growth in two pulmonary metastases tumor models and significantly reduced the dendritic and NK cell pools residing in lymphoid organs in a B7-H1-dependent manner. Neutralization of IL-18 by RNA interference in tumors or systemically by IL-18-binding protein dramatically reduced the accumulation of Kit(+)CD11b(-) NK cells in tumor bearers. Together, our findings show that IL-18 produced by tumor cells elicits Kit(+)CD11b(-) NK cells endowed with B7-H1-dependent immunoablative functions in mice.


Assuntos
Interleucina-18/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Animais , Antígeno B7-H1/análise , Antígeno B7-H1/fisiologia , Antígeno CD11b/análise , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-kit/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA