Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 8(84): eade7652, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37327321

RESUMO

NLRP3 inflammasome activation is a highly regulated process for controlling secretion of the potent inflammatory cytokines IL-1ß and IL-18 that are essential during bacterial infection, sterile inflammation, and disease, including colitis, diabetes, Alzheimer's disease, and atherosclerosis. Diverse stimuli activate the NLRP3 inflammasome, and unifying upstream signals has been challenging to identify. Here, we report that a common upstream step in NLRP3 inflammasome activation is the dissociation of the glycolytic enzyme hexokinase 2 from the voltage-dependent anion channel (VDAC) in the outer membrane of mitochondria. Hexokinase 2 dissociation from VDAC triggers activation of inositol triphosphate receptors, leading to release of calcium from the ER, which is taken up by mitochondria. This influx of calcium into mitochondria leads to oligomerization of VDAC, which is known to form a macromolecule-sized pore in the outer membranes of mitochondria that allows proteins and mitochondrial DNA (mtDNA), often associated with apoptosis and inflammation, respectively, to exit the mitochondria. We observe that VDAC oligomers aggregate with NLRP3 during initial assembly of the multiprotein oligomeric NLRP3 inflammasome complex. We also find that mtDNA is necessary for NLRP3 association with VDAC oligomers. These data, together with other recent work, help to paint a more complete picture of the pathway leading to NLRP3 inflammasome activation.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Hexoquinase/metabolismo , Cálcio/metabolismo , Mitocôndrias/metabolismo , Canais de Ânion Dependentes de Voltagem/metabolismo , DNA Mitocondrial/metabolismo , Inflamação/metabolismo
2.
Immunometabolism (Cobham) ; 5(2): e00024, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37128291

RESUMO

Bacterial cell wall peptidoglycan is composed of innate immune ligands and, due to its important structural role, also regulates access to many other innate immune ligands contained within the bacteria. There is a growing body of literature demonstrating how innate immune recognition impacts the metabolic functions of immune cells and how metabolic changes are not only important to inflammatory responses but are often essential. Peptidoglycan is primarily sensed in the context of the whole bacteria during lysosomal degradation; consequently, the innate immune receptors for peptidoglycan are primarily intracellular cytosolic innate immune sensors. However, during bacterial growth, peptidoglycan fragments are shed and can be found in the bloodstream of humans and mice, not only during infection but also derived from the abundant bacterial component of the gut microbiota. These peptidoglycan fragments influence cells throughout the body and are important for regulating inflammation and whole-body metabolic function. Therefore, it is important to understand how peptidoglycan-induced signals in innate immune cells and cells throughout the body interact to regulate how the body responds to both pathogenic and nonpathogenic bacteria. This mini-review will highlight key research regarding how cellular metabolism shifts in response to peptidoglycan and how systemic peptidoglycan sensing impacts whole-body metabolic function.

3.
Gut ; 72(11): 2068-2080, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37080587

RESUMO

OBJECTIVE: Perianal Crohn's disease (pCD) occurs in up to 40% of patients with CD and is associated with poor quality of life, limited treatment responses and poorly understood aetiology. We performed a genetic association study comparing CD subjects with and without perianal disease and subsequently performed functional follow-up studies for a pCD associated SNP in Complement Factor B (CFB). DESIGN: Immunochip-based meta-analysis on 4056 pCD and 11 088 patients with CD from three independent cohorts was performed. Serological and clinical variables were analysed by regression analyses. Risk allele of rs4151651 was introduced into human CFB plasmid by site-directed mutagenesis. Binding of recombinant G252 or S252 CFB to C3b and its cleavage was determined in cell-free assays. Macrophage phagocytosis in presence of recombinant CFB or serum from CFB risk, or protective CD or healthy subjects was assessed by flow cytometry. RESULTS: Perianal complications were associated with colonic involvement, OmpC and ASCA serology, and serology quartile sum score. We identified a genetic association for pCD (rs4151651), a non-synonymous SNP (G252S) in CFB, in all three cohorts. Recombinant S252 CFB had reduced binding to C3b, its cleavage was impaired, and complement-driven phagocytosis and cytokine secretion were reduced compared with G252 CFB. Serine 252 generates a de novo glycosylation site in CFB. Serum from homozygous risk patients displayed significantly decreased macrophage phagocytosis compared with non-risk serum. CONCLUSION: pCD-associated rs4151651 in CFB is a loss-of-function mutation that impairs its cleavage, activation of alternative complement pathway, and pathogen phagocytosis thus implicating the alternative complement pathway and CFB in pCD aetiology.


Assuntos
Fator B do Complemento , Doença de Crohn , Humanos , Fator B do Complemento/genética , Doença de Crohn/complicações , Qualidade de Vida , Seguimentos , Fagocitose
4.
J Leukoc Biol ; 109(1): 161-172, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32941658

RESUMO

Malassezia spp. are common eukaryotic yeasts that colonize mammalian skin. Recently, the authors and others have observed that Malassezia globosa and Malassezia restricta can be found in the intestines in the context of certain diseases, including Crohn's disease and pancreatic cancer. In order to better understand the nature of innate inflammatory responses to these yeasts, inflammatory responses induced by M. restricta and M. globosa in mouse bone marrow-derived Mϕs (BMDM) and dendritic cells (BMDC) are evaluated. While Malassezia yeasts induce proinflammatory cytokine production from both Mϕs and dendritic cells, the levels of production from BMDC were more pronounced. Both M. restricta and M. globosa activated inflammatory cytokine production from BMDC in large part through Dectin2 and CARD9 signaling, although additional receptors appear to be involved in phagocytosis and activation of reactive oxygen production in response to the yeasts. Both M. restricta and M. globosa stimulate production of pro-IL-1ß as well as activation of the NLRP3 inflammasome. NLRP3 inflammasome activation by Malassezia fungi requires SYK signaling, potassium efflux and actin rearrangement. Together, the data further the understanding of the coordinated involvement of multiple innate immune receptors in recognizing Malassezia globosa and Malassezia restricta and orchestrating phagocyte inflammatory and antimicrobial responses.


Assuntos
Inflamassomos/imunologia , Inflamação/imunologia , Malassezia/imunologia , Micoses/imunologia , Fagócitos/imunologia , Animais , Citocinas/imunologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
5.
Cell Host Microbe ; 25(3): 377-388.e6, 2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30850233

RESUMO

Inflammatory bowel disease (IBD) is characterized by alterations in the intestinal microbiota and altered immune responses to gut microbiota. Evidence is accumulating that IBD is influenced by not only commensal bacteria but also commensal fungi. We characterized fungi directly associated with the intestinal mucosa in healthy people and Crohn's disease patients and identified fungi specifically abundant in patients. One of these, the common skin resident fungus Malassezia restricta, is also linked to the presence of an IBD-associated polymorphism in the gene for CARD9, a signaling adaptor important for anti-fungal defense. M. restricta elicits innate inflammatory responses largely through CARD9 and is recognized by Crohn's disease patient anti-fungal antibodies. This yeast elicits strong inflammatory cytokine production from innate cells harboring the IBD-linked polymorphism in CARD9 and exacerbates colitis via CARD9 in mouse models of disease. Collectively, these results suggest that targeting specific commensal fungi may be a therapeutic strategy for IBD.


Assuntos
Colite/patologia , Colite/fisiopatologia , Doença de Crohn/patologia , Doença de Crohn/fisiopatologia , Trato Gastrointestinal/microbiologia , Malassezia/crescimento & desenvolvimento , Malassezia/isolamento & purificação , Animais , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Camundongos
6.
Nat Rev Immunol ; 18(4): 243-254, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29292393

RESUMO

The innate immune system recognizes microbial products using germline-encoded receptors that initiate inflammatory responses to infection. The bacterial cell wall component peptidoglycan is a prime example of a conserved pathogen-associated molecular pattern (PAMP) for which the innate immune system has evolved sensing mechanisms. Peptidoglycan is a direct target for innate immune receptors and also regulates the accessibility of other PAMPs to additional innate immune receptors. Subtle structural modifications to peptidoglycan can influence the ability of the innate immune system to detect bacteria and can allow bacteria to evade or alter host defences. This Review focuses on the mechanisms of peptidoglycan recognition that are used by mammalian cells and discusses new insights into the role of peptidoglycan recognition in inflammation, metabolism, immune homeostasis and disease.


Assuntos
Imunidade Inata , Peptidoglicano/imunologia , Animais , Proteínas de Transporte/imunologia , Microbioma Gastrointestinal/imunologia , Humanos , Imunidade nas Mucosas , Modelos Imunológicos , Proteínas NLR/imunologia , Peptidoglicano/química , Peptidoglicano/metabolismo , Receptores de Superfície Celular/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia
7.
Cell Host Microbe ; 22(4): 543-551.e4, 2017 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-28943328

RESUMO

Humans do not usually develop effective immunity to Staphylococcus aureus reinfection. Using a murine model that mimics human infection, we show that lack of protective immunity to S. aureus systemic reinfection is associated with robust interleukin-10 (IL-10) production and impaired protective Th17 responses. In dendritic cell co-culture assays, priming with S. aureus promotes robust T cell proliferation, but limits Th cells polarization and production of IL-1ß and other cytokines important for Th1 and Th17 differentiation. We show that O-acetylation of peptidoglycan, a mechanism utilized by S. aureus to block bacterial cell wall breakdown, limits the induction of pro-inflammatory signals required for optimal Th17 polarization. IL-10 deficiency in mice restores protective immunity to S. aureus infection, and adjuvancy with a staphylococcal peptidoglycan O-acetyltransferase mutant reduces IL-10, increases IL-1ß, and promotes development of IL-17-dependent, Th cell-transferable protective immunity. Overall, our study suggests a mechanism whereby S. aureus modulates cytokines critical for induction of protective Th17 immunity.


Assuntos
Acetiltransferases/imunologia , Peptidoglicano/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Células Th17/imunologia , Acetilação , Acetiltransferases/metabolismo , Imunidade Adaptativa , Animais , Técnicas de Cocultura , Células Dendríticas/imunologia , Feminino , Humanos , Interleucina-10/imunologia , Interleucina-1beta/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptidoglicano/metabolismo
8.
J Immunol ; 198(10): 4036-4045, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28411186

RESUMO

Type I IFNs are a cytokine family essential for antiviral defense. More recently, type I IFNs were shown to be important during bacterial infections. In this article, we show that, in addition to known cytokine functions, IFN-ß is antimicrobial. Parts of the IFN-ß molecular surface (especially helix 4) are cationic and amphipathic, both classic characteristics of antimicrobial peptides, and we observed that IFN-ß can directly kill Staphylococcus aureus Further, a mutant S. aureus that is more sensitive to antimicrobial peptides was killed more efficiently by IFN-ß than was the wild-type S. aureus, and immunoblotting showed that IFN-ß interacts with the bacterial cell surface. To determine whether specific parts of IFN-ß are antimicrobial, we synthesized IFN-ß helix 4 and found that it is sufficient to permeate model prokaryotic membranes using synchrotron x-ray diffraction and that it is sufficient to kill S. aureus These results suggest that, in addition to its well-known signaling activity, IFN-ß may be directly antimicrobial and be part of a growing family of cytokines and chemokines, called kinocidins, that also have antimicrobial properties.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Interferon beta/fisiologia , Staphylococcus aureus/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Humanos , Interferon beta/química , Interferon beta/metabolismo , Interferon beta/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Difração de Raios X
9.
J Leukoc Biol ; 101(1): 127-134, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27576461

RESUMO

Antibiotics have proven to be enormously effective tools in combating infectious diseases. A common roadblock to the effective use of antibiotics is the development of antibiotic resistance. We have recently observed that the very mechanism by which methicillin-resistant Staphylococcus aureus (MRSA) becomes antibiotic resistant causes the organism to be more inflammatory to innate immune cells. In this review, we offer some thoughts on the ways in which antibiotics have been observed to influence immune responses to bacteria.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Inflamação/patologia , Animais , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos
10.
Cell ; 166(3): 624-636, 2016 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-27374331

RESUMO

Degradation of Gram-positive bacterial cell wall peptidoglycan in macrophage and dendritic cell phagosomes leads to activation of the NLRP3 inflammasome, a cytosolic complex that regulates processing and secretion of interleukin (IL)-1ß and IL-18. While many inflammatory responses to peptidoglycan are mediated by detection of its muramyl dipeptide component in the cytosol by NOD2, we report here that NLRP3 inflammasome activation is caused by release of N-acetylglucosamine that is detected in the cytosol by the glycolytic enzyme hexokinase. Inhibition of hexokinase by N-acetylglucosamine causes its dissociation from mitochondria outer membranes, and we found that this is sufficient to activate the NLRP3 inflammasome. In addition, we observed that glycolytic inhibitors and metabolic conditions affecting hexokinase function and localization induce inflammasome activation. While previous studies have demonstrated that signaling by pattern recognition receptors can regulate metabolic processes, this study shows that a metabolic enzyme can act as a pattern recognition receptor. PAPERCLIP.


Assuntos
Hexoquinase/metabolismo , Inflamassomos/metabolismo , Peptidoglicano/metabolismo , Receptores Imunológicos/metabolismo , Acetilação , Acetilglucosamina/metabolismo , Animais , Bacillus anthracis/metabolismo , Parede Celular/metabolismo , Células Dendríticas/metabolismo , Glicólise , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Modelos Biológicos , Monócitos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Potássio/metabolismo
11.
Cell Host Microbe ; 18(5): 604-12, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26567511

RESUMO

Methicillin-resistant S. aureus (MRSA) is a leading health problem. Compared to methicillin-sensitive S. aureus, MRSA infections are associated with greater morbidity and mortality, but the mechanisms underlying MRSA pathogenicity are unclear. Here we show that the protein conferring ß-lactam antibiotic resistance, penicillin-binding protein 2A (encoded by the mecA gene), directly contributes to pathogenicity during MRSA infection. MecA induction leads to a reduction in peptidoglycan cross-linking that allows for enhanced degradation and detection by phagocytes, resulting in robust IL-1ß production. Peptidoglycan isolated from ß-lactam-challenged MRSA strongly induces the NLRP3 inflammasome in macrophages, but these effects are lost upon peptidoglycan solubilization. Mutant MRSA bacteria with naturally occurring reduced peptidoglycan cross-links induce high IL-1ß levels in vitro and cause increased pathology in vivo. ß-lactam treatment of MRSA skin infection exacerbates immunopathology, which is IL-1 dependent. Thus, antibiotic-induced expression of mecA during MRSA skin infection contributes to immunopathology by altering peptidoglycan structure.


Assuntos
Proteínas de Bactérias/metabolismo , Interleucina-1beta/metabolismo , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Proteínas de Ligação às Penicilinas/metabolismo , Peptidoglicano/metabolismo , Infecções Estafilocócicas/microbiologia , Animais , Inflamassomos/imunologia , Inflamassomos/metabolismo , Lactamas/uso terapêutico , Staphylococcus aureus Resistente à Meticilina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/imunologia , Virulência
12.
J Immunol ; 189(9): 4537-45, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23008447

RESUMO

The importance of type I IFNs in the host response to viral infection is well established; however, their role in bacterial infection is not fully understood. Several bacteria (both Gram-positive and -negative) have been shown to induce IFN-ß production in myeloid cells, but this IFN-ß is not always beneficial to the host. We examined whether Staphylococcus aureus induces IFN-ß from myeloid phagocytes, and if so, whether it is helpful or harmful to the host to do so. We found that S. aureus poorly induces IFN-ß production compared with other bacteria. S. aureus is highly resistant to degradation in the phagosome because it is resistant to lysozyme. Using a mutant that is more sensitive to lysozyme, we show that phagosomal degradation and release of intracellular ligands is essential for induction of IFN-ß and inflammatory chemokines downstream of IFN-ß. Further, we found that adding exogenous IFN-ß during S. aureus infection (in vitro and in vivo) was protective. Together, the data demonstrate that failure to induce IFN-ß production during S. aureus infection contributes to pathogenicity.


Assuntos
Interferon beta , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Interferon beta/antagonistas & inibidores , Interferon beta/biossíntese , Interferon beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/imunologia , Infecções Estafilocócicas/sangue , Staphylococcus aureus/genética
13.
J Clin Invest ; 122(9): 3316-29, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22922257

RESUMO

The myeloid-specific transcription factor, CCAAT/enhancer-binding protein ε (C/EBPε) is a critical mediator of myelopoiesis. Mutation of this gene is responsible for neutrophil-specific granule deficiency in humans, a condition that confers susceptibility to Staphylococcus aureus infection. We found that C/EBPε-deficient mice are severely affected by infection with S. aureus, and C/EBPε deficiency in neutrophils contributes to the infectious phenotype. Conversely, exposure to the epigenetic modulator nicotinamide (vitamin B3) increased expression of C/EBPε in WT myeloid cells. Further, nicotinamide increased the activity of C/EBPε and select downstream antimicrobial targets, particularly in neutrophils. In a systemic murine infection model as well as in murine and human peripheral blood, nicotinamide enhanced killing of S. aureus by up to 1,000 fold but had no effect when administered to either C/EBPε-deficient mice or mice depleted of neutrophils. Nicotinamide was efficacious in both prophylactic and therapeutic settings. Our findings suggest that C/EBPε is an important target to boost killing of bacteria by the innate immune system.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Niacinamida/farmacologia , Infecções Cutâneas Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/fisiologia , Acetilação , Animais , Antibacterianos/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica , Histonas/metabolismo , Humanos , Imunidade Inata , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Viabilidade Microbiana , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/microbiologia , Niacinamida/fisiologia , Regiões Promotoras Genéticas , Infecções Cutâneas Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/patologia
14.
J Antimicrob Chemother ; 67(9): 2191-4, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22661572

RESUMO

OBJECTIVES: Methicillin-resistant Staphylococcus aureus (MRSA) poses a major problem to public health worldwide. MRSA strains with increased resistance to vancomycin cause infections that are associated with greater morbidity and threaten the use of this once gold-standard antistaphylococcal drug. We investigated whether encapsulation of vancomycin within liposomes could improve its antistaphylococcal activity. METHODS: Two liposomal formulations of vancomycin were prepared using a rehydration-dehydration method. MICs and MBCs of the liposomal vancomycin for strains of MRSA were determined. The efficacy of one of the liposomal vancomycin formulations was also investigated in a time-kill assay in vitro and in a murine systemic infection model. RESULTS: Encapsulation in either liposome preparation decreased the vancomycin MICs and MBCs for MRSA strains by approximately 2-fold. Liposomal vancomycin increased killing of MRSA in vitro in a kinetic study. In a systemic murine infection model, treatment with a 50 mg/kg intraperitoneal injection of liposomal vancomycin improved kidney clearance of a USA300 strain by 1 log compared with an injection of 50 mg/kg of free vancomycin. CONCLUSIONS: Our findings suggest that entrapment within liposomes could improve the antistaphylococcal efficacy of vancomycin.


Assuntos
Antibacterianos/administração & dosagem , Lipossomos/administração & dosagem , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Vancomicina/administração & dosagem , Animais , Antibacterianos/farmacocinética , Bacteriemia/tratamento farmacológico , Bacteriemia/microbiologia , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacocinética , Lipossomos/farmacocinética , Masculino , Camundongos , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Infecções Estafilocócicas/microbiologia , Resultado do Tratamento , Vancomicina/farmacocinética
15.
J Immunol ; 188(12): 6205-15, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22566567

RESUMO

Although the protective role of type II IFN, or IFN-γ, against Mycobacterium tuberculosis has been established, the effects of type I IFNs are still unclear. One potential confounding factor is the overlap of function between the two signaling pathways. We used mice carrying null mutations in the type I IFNR, type II IFNR, or both and compared their immune responses to those of wild-type mice following aerosol infection with M. tuberculosis. We discovered that, in the absence of a response to IFN-γ, type I IFNs play a nonredundant protective role against tuberculosis. Mice unable to respond to both types of IFNs had more severe lung histopathology for similar bacterial loads and died significantly earlier than did mice with impaired IFN-γ signaling alone. We excluded a role for type I IFN in T cell recruitment, which was IFN-γ dependent, whereas both types of IFNs were required for optimal NK cell recruitment to the lungs. Type I IFN had a time-dependent influence on the composition of lung myeloid cell populations, in particular by limiting the abundance of M. tuberculosis-infected recruited macrophages after the onset of adaptive immunity. We confirmed that response to IFN-γ was essential to control intracellular mycobacterial growth, without any additional effect of type I IFN. Together, our results imply a model in which type I IFN limit the number of target cells that M. tuberculosis can infect in the lungs, whereas IFN-γ enhances their ability to restrict bacterial growth.


Assuntos
Interferon Tipo I/imunologia , Interferon gama/imunologia , Tuberculose Pulmonar/imunologia , Animais , Citocinas/biossíntese , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
16.
Immunity ; 36(3): 401-14, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22342844

RESUMO

We report that in the presence of signal 1 (NF-κB), the NLRP3 inflammasome was activated by mitochondrial apoptotic signaling that licensed production of interleukin-1ß (IL-1ß). NLRP3 secondary signal activators such as ATP induced mitochondrial dysfunction and apoptosis, resulting in release of oxidized mitochondrial DNA (mtDNA) into the cytosol, where it bound to and activated the NLRP3 inflammasome. The antiapoptotic protein Bcl-2 inversely regulated mitochondrial dysfunction and NLRP3 inflammasome activation. Mitochondrial DNA directly induced NLRP3 inflammasome activation, because macrophages lacking mtDNA had severely attenuated IL-1ß production, yet still underwent apoptosis. Both binding of oxidized mtDNA to the NLRP3 inflammasome and IL-1ß secretion could be competitively inhibited by the oxidized nucleoside 8-OH-dG. Thus, our data reveal that oxidized mtDNA released during programmed cell death causes activation of the NLRP3 inflammasome. These results provide a missing link between apoptosis and inflammasome activation, via binding of cytosolic oxidized mtDNA to the NLRP3 inflammasome.


Assuntos
Apoptose/imunologia , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , DNA Mitocondrial/imunologia , DNA Mitocondrial/metabolismo , Inflamassomos/imunologia , Inflamassomos/metabolismo , Animais , Expressão Gênica , Interleucina-1beta/biossíntese , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Oxirredução , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Transdução de Sinais
17.
J Immunol ; 187(11): 6002-10, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22031762

RESUMO

Signaling by innate immune receptors initiates and orchestrates the overall immune responses to infection. Macrophage receptors recognizing pathogens can be broadly grouped into surface receptors and receptors restricted to intracellular compartments, such as phagosomes and the cytoplasm. There is an expectation that ingestion and degradation of microorganisms by phagocytes contributes to activation of intracellular innate receptors, although direct demonstrations of this are rare, and many model ligands are studied in soluble form, outside of their microbial context. By comparing a wild-type strain of Staphylococcus aureus and a lysozyme-sensitive mutant, we have been able directly to address the role of degradation of live bacteria by mouse macrophages in determining the overall innate cellular inflammatory response. Our investigations revealed a biphasic response to S. aureus that consisted of an initial signal resulting from the engagement of surface TLR2, followed by a later, second wave on inflammatory gene induction. This second wave of inflammatory signaling was dependent on and correlated with the timing of bacterial degradation in phagosomes. We found that TLR2 signaling followed by TLR2/TLR9 signaling enhanced sensitivity to small numbers of bacteria. We further found that treating wild-type bacteria with the peptidoglycan synthesis-inhibiting antibiotic vancomycin made S. aureus more susceptible to degradation and resulted in increased inflammatory responses, similar to those observed for mutant degradation-sensitive bacteria.


Assuntos
Macrófagos/imunologia , Fagocitose/imunologia , Fagossomos/imunologia , Infecções Estafilocócicas/imunologia , Receptores Toll-Like/imunologia , Animais , Imunidade Inata/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Ligantes , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Fagossomos/metabolismo , Reação em Cadeia da Polimerase , Transdução de Sinais/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/imunologia , Receptores Toll-Like/metabolismo
18.
PLoS Pathog ; 7(5): e1002063, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21637811

RESUMO

Adaptive immunity to Mycobacterium tuberculosis controls progressive bacterial growth and disease but does not eradicate infection. Among CD4+ T cells in the lungs of M. tuberculosis-infected mice, we observed that few produced IFN-γ without ex vivo restimulation. Therefore, we hypothesized that one mechanism whereby M. tuberculosis avoids elimination is by limiting activation of CD4+ effector T cells at the site of infection in the lungs. To test this hypothesis, we adoptively transferred Th1-polarized CD4+ effector T cells specific for M. tuberculosis Ag85B peptide 25 (P25TCRTh1 cells), which trafficked to the lungs of infected mice and exhibited antigen-dependent IFN-γ production. During the early phase of infection, ∼10% of P25TCRTh1 cells produced IFN-γ in vivo; this declined to <1% as infection progressed to chronic phase. Bacterial downregulation of fbpB (encoding Ag85B) contributed to the decrease in effector T cell activation in the lungs, as a strain of M. tuberculosis engineered to express fbpB in the chronic phase stimulated P25TCRTh1 effector cells at higher frequencies in vivo, and this resulted in CD4+ T cell-dependent reduction of lung bacterial burdens and prolonged survival of mice. Administration of synthetic peptide 25 alone also increased activation of endogenous antigen-specific effector cells and reduced the bacterial burden in the lungs without apparent host toxicity. These results indicate that CD4+ effector T cells are activated at suboptimal frequencies in tuberculosis, and that increasing effector T cell activation in the lungs by providing one or more epitope peptides may be a successful strategy for TB therapy.


Assuntos
Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/patologia , Proliferação de Células , Mycobacterium tuberculosis/patogenicidade , Tuberculose/microbiologia , Tuberculose/patologia , Animais , Antígenos de Bactérias/metabolismo , Antígenos CD4/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Interferon gama/metabolismo , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Viabilidade Microbiana , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/fisiologia , Tuberculose/metabolismo
19.
Nature ; 472(7344): 471-5, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21525931

RESUMO

Innate immune cells must be able to distinguish between direct binding to microbes and detection of components shed from the surface of microbes located at a distance. Dectin-1 (also known as CLEC7A) is a pattern-recognition receptor expressed by myeloid phagocytes (macrophages, dendritic cells and neutrophils) that detects ß-glucans in fungal cell walls and triggers direct cellular antimicrobial activity, including phagocytosis and production of reactive oxygen species (ROS). In contrast to inflammatory responses stimulated upon detection of soluble ligands by other pattern-recognition receptors, such as Toll-like receptors (TLRs), these responses are only useful when a cell comes into direct contact with a microbe and must not be spuriously activated by soluble stimuli. In this study we show that, despite its ability to bind both soluble and particulate ß-glucan polymers, Dectin-1 signalling is only activated by particulate ß-glucans, which cluster the receptor in synapse-like structures from which regulatory tyrosine phosphatases CD45 and CD148 (also known as PTPRC and PTPRJ, respectively) are excluded (Supplementary Fig. 1). The 'phagocytic synapse' now provides a model mechanism by which innate immune receptors can distinguish direct microbial contact from detection of microbes at a distance, thereby initiating direct cellular antimicrobial responses only when they are required.


Assuntos
Imunidade Inata/imunologia , Sinapses Imunológicas/imunologia , Proteínas de Membrana/imunologia , Modelos Imunológicos , Proteínas do Tecido Nervoso/imunologia , Fagocitose/imunologia , Animais , Parede Celular/química , Parede Celular/imunologia , Células Cultivadas , Humanos , Lectinas Tipo C , Antígenos Comuns de Leucócito/deficiência , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/imunologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/deficiência , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/imunologia , Transdução de Sinais/imunologia , Solubilidade , beta-Glucanas/química , beta-Glucanas/imunologia
20.
Cell Host Microbe ; 7(1): 38-49, 2010 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-20114027

RESUMO

IL-1beta produced by phagocytes is important for protection against the mucosal pathogen Staphylococcus aureus. Processing and maturation of this cytokine requires activation of the multiprotein inflammasome complex. We observed that the bacterial cell wall component peptidoglycan (PGN) must be particulate and internalized via phagocytosis to activate NLRP3 inflammasomes and IL-1beta secretion. In the context of S. aureus infection of macrophages, we find that phagocytosis and lysozyme-based bacterial cell wall degradation are necessary to induce IL-1beta secretion. Further, an S. aureus enzyme, PGN O-acetyltransferase A, previously demonstrated to make cell wall PGN resistant to lysozyme, strongly suppresses inflammasome activation and inflammation in vitro and in vivo. These observations demonstrate that phagocytosis and lysozyme-based cell wall degradation of S. aureus are functionally coupled to inflammasome activation and IL-1beta secretion and illustrate a case whereby a bacterium specifically subverts IL-1beta secretion through chemical modification of its cell wall PGN.


Assuntos
Proteínas de Transporte/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Muramidase/metabolismo , Peptidoglicano/metabolismo , Fagossomos/enzimologia , Staphylococcus aureus/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Biológicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Staphylococcus aureus/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA