Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Front Immunol ; 14: 1224383, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38146368

RESUMO

Chronic obstructive pulmonary disease (COPD) is a major health issue primarily caused by cigarette smoke (CS) and characterized by breathlessness and repeated airway inflammation. NLRP6 is a cytosolic innate receptor controlling intestinal inflammation and orchestrating the colonic host-microbial interface. However, its roles in the lungs remain largely unexplored. Using CS exposure models, our data show that airway inflammation is strongly impaired in Nlrp6-deficient mice with drastically fewer recruited neutrophils, a key cell subset in inflammation and COPD. We found that NLRP6 expression in lung epithelial cells is important to control airway and lung tissue inflammation in an inflammasome-dependent manner. Since gut-derived metabolites regulate NLRP6 inflammasome activation in intestinal epithelial cells, we investigated the link between NLRP6, CS-driven lung inflammation, and gut microbiota composition. We report that acute CS exposure alters gut microbiota in both wild-type (WT) and Nlrp6-deficient mice and that antibiotic treatment decreases CS-induced lung inflammation. In addition, gut microbiota transfer from dysbiotic Nlrp6-deficient mice to WT mice decreased airway lung inflammation in WT mice, highlighting an NLRP6-dependent gut-to-lung axis controlling pulmonary inflammation.


Assuntos
Microbioma Gastrointestinal , Pneumonia , Receptores de Superfície Celular , Poluição por Fumaça de Tabaco , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/microbiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/patologia , Fezes/microbiologia , Bactérias/classificação , Bactérias/metabolismo , Biodiversidade , Expressão Gênica
2.
Trends Immunol ; 44(12): 1014-1030, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37951789

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a fatal chronic interstitial lung disease (ILD) that affects lung mechanical functions and gas exchange. IPF is caused by increased fibroblast activity and collagen deposition that compromise the alveolar-capillary barrier. Identifying an effective therapy for IPF remains a clinical challenge. Chemokines are key proteins in cell communication that have functions in immunity as well as in tissue homeostasis, damage, and repair. Chemokine receptor signaling induces the activation and proliferation of lung-resident cells, including alveolar macrophages (AMs) and fibroblasts. AMs are an important source of chemokines and cytokines during IPF. We highlight the complexity of this system and, based on insights from genetic and transcriptomic studies, propose a new role for homeostatic chemokine imbalance in IPF, with implications for putative therapeutic targets.


Assuntos
Fibrose Pulmonar Idiopática , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/etiologia , Fibrose Pulmonar Idiopática/metabolismo , Quimiocinas/metabolismo , Macrófagos Alveolares , Citocinas/metabolismo , Transdução de Sinais , Pulmão
3.
Arch Toxicol ; 97(4): 1001-1015, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36840754

RESUMO

IL-1α is an intracellular danger signal (DAMP) released by macrophages contributing to the development of silica-induced lung inflammation. The exact molecular mechanism orchestrating IL-1α extracellular release from particle-exposed macrophages is still unclear. To delineate this process, murine J774 and bone-marrow derived macrophages were exposed to increasing concentrations (1-40 cm2/ml) of a set of amorphous and crystalline silica particles with different surface chemical features. In particular, these characteristics include the content of nearly free silanols (NFS), a silanol population responsible for silica cytotoxicity recently identified. We first observed de novo stocks of IL-1α in macrophages after silica internalization regardless of particle physico-chemical characteristics and cell stress. IL-1α intracellular production and accumulation were observed by exposing macrophages to biologically-inert or cytotoxic crystalline and amorphous silicas. In contrast, only NFS-rich reactive silica particles triggered IL-1α release into the extracellular milieu from necrotic macrophages. We demonstrate that IL-1α is actively secreted through the formation of gasdermin D (GSDMD) pores in the plasma membrane and not passively released after macrophage plasma membrane lysis. Our findings indicate that the GSDMD pore-dependent secretion of IL-1α stock from macrophages solely depends on cytotoxicity induced by NFS-rich silica. This new regulated process represents a key first event in the mechanism of silica toxicity, suitable to refine the existing adverse outcome pathway (AOP) for predicting the inflammatory activity of silicas.


Assuntos
Gasderminas , Macrófagos , Camundongos , Animais , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Necrose , Dióxido de Silício/química
4.
Front Immunol ; 13: 918507, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36045672

RESUMO

Chronic pulmonary inflammation and chronic obstructive pulmonary disease (COPD) are major health issues largely due to air pollution and cigarette smoke (CS) exposure. The role of the innate receptor NLRP3 (nucleotide-binding domain and leucine-rich repeat containing protein 3) orchestrating inflammation through formation of an inflammasome complex in CS-induced inflammation or COPD remains controversial. Using acute and subchronic CS exposure models, we found that Nlrp3-deficient mice or wild-type mice treated with the NLRP3 inhibitor MCC950 presented an important reduction of inflammatory cells recruited into the bronchoalveolar space and of pulmonary inflammation with decreased chemokines and cytokines production, in particular IL-1ß demonstrating the key role of NLRP3. Furthermore, mice deficient for Caspase-1/Caspase-11 presented also decreased inflammation parameters, suggesting a role for the NLRP3 inflammasome. Importantly we showed that acute CS-exposure promotes NLRP3-dependent cleavage of gasdermin D in macrophages present in the bronchoalveolar space and in bronchial airway epithelial cells. Finally, Gsdmd-deficiency reduced acute CS-induced lung and bronchoalveolar space inflammation and IL-1ß secretion. Thus, we demonstrated in our model that NLRP3 and gasdermin D are key players in CS-induced pulmonary inflammation and IL-1ß release potentially through gasdermin D forming-pore and/or pyroptoctic cell death.


Assuntos
Fumar Cigarros , Pneumonia , Doença Pulmonar Obstrutiva Crônica , Animais , Caspase 1/metabolismo , Fumar Cigarros/efeitos adversos , Células Epiteliais/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pneumonia/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Nicotiana/metabolismo
5.
Cell Death Dis ; 13(3): 269, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35338116

RESUMO

Stimulator of interferon genes (STING) contributes to immune responses against tumors and may control viral infection including SARS-CoV-2 infection. However, activation of the STING pathway by airway silica or smoke exposure leads to cell death, self-dsDNA release, and STING/type I IFN dependent acute lung inflammation/ARDS. The inflammatory response induced by a synthetic non-nucleotide-based diABZI STING agonist, in comparison to the natural cyclic dinucleotide cGAMP, is unknown. A low dose of diABZI (1 µg by endotracheal route for 3 consecutive days) triggered an acute neutrophilic inflammation, disruption of the respiratory barrier, DNA release with NET formation, PANoptosis cell death, and inflammatory cytokines with type I IFN dependent acute lung inflammation. Downstream upregulation of DNA sensors including cGAS, DDX41, IFI204, as well as NLRP3 and AIM2 inflammasomes, suggested a secondary inflammatory response to dsDNA as a danger signal. DNase I treatment, inhibition of NET formation together with an investigation in gene-deficient mice highlighted extracellular DNA and TLR9, but not cGAS, as central to diABZI-induced neutrophilic response. Therefore, activation of acute cell death with DNA release may lead to ARDS which may be modeled by diABZI. These results show that airway targeting by STING activator as a therapeutic strategy for infection may enhance lung inflammation with severe ARDS. STING agonist diABZI induces neutrophilic lung inflammation and PANoptosis A, Airway STING priming induce a neutrophilic lung inflammation with epithelial barrier damage, double-stranded DNA release in the bronchoalvelolar space, cell death, NETosis and type I interferon release. B, 1. The diamidobenzimidazole (diABZI), a STING agonist is internalized into the cytoplasm through unknown receptor and induce the activation and dimerization of STING followed by TBK1/IRF3 phosporylation leading to type I IFN response. STING activation also leads to NF-kB activation and the production of pro-inflammatory cytokines TNFα and IL-6. 2. The activation of TNFR1 and IFNAR1 signaling pathway results in ZBP1 and RIPK3/ASC/CASP8 activation leading to MLKL phosphorylation and necroptosis induction. 3. This can also leads to Caspase-3 cleavage and apoptosis induction. 4. Self-dsDNA or mtDNA sensing by NLRP3 or AIM2 induces inflammsome formation leading to Gasdermin D cleavage enabling Gasdermin D pore formation and the release mature IL-1ß and pyroptosis. NLRP3 inflammasome formation can be enhanced by the ZBP1/RIPK3/CASP8 complex. 5. A second signal of STING activation with diABZI induces cell death and the release of self-DNA which is sensed by cGAS and form 2'3'-cGAMP leading to STING hyper activation, the amplification of TBK1/IRF3 and NF-kB pathway and the subsequent production of IFN-I and inflammatory TNFα and IL-6. This also leads to IFI204 and DDX41 upregulation thus, amplifying the inflammatory loop. The upregulation of apoptosis, pyroptosis and necroptosis is indicative of STING-dependent PANoptosis.


Assuntos
COVID-19 , Síndrome do Desconforto Respiratório , Animais , Citocinas/metabolismo , DNA , Inflamassomos/metabolismo , Interleucina-6/metabolismo , Camundongos , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Proteínas de Ligação a RNA , Síndrome do Desconforto Respiratório/genética , SARS-CoV-2 , Fator de Necrose Tumoral alfa/metabolismo
6.
Immunity ; 54(9): 2024-2041.e8, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34473957

RESUMO

Sepsis results in elevated adenosine in circulation. Extracellular adenosine triggers immunosuppressive signaling via the A2a receptor (A2aR). Sepsis survivors develop persistent immunosuppression with increased risk of recurrent infections. We utilized the cecal ligation and puncture (CLP) model of sepsis and subsequent infection to assess the role of adenosine in post-sepsis immune suppression. A2aR-deficient mice showed improved resistance to post-sepsis infections. Sepsis expanded a subset of CD39hi B cells and elevated extracellular adenosine, which was absent in mice lacking CD39-expressing B cells. Sepsis-surviving B cell-deficient mice were more resistant to secondary infections. Mechanistically, metabolic reprogramming of septic B cells increased production of ATP, which was converted into adenosine by CD39 on plasmablasts. Adenosine signaling via A2aR impaired macrophage bactericidal activity and enhanced interleukin-10 production. Septic individuals exhibited expanded CD39hi plasmablasts and adenosine accumulation. Our study reveals CD39hi plasmablasts and adenosine as important drivers of sepsis-induced immunosuppression with relevance in human disease.


Assuntos
Adenosina/imunologia , Antígenos CD/imunologia , Apirase/imunologia , Tolerância Imunológica/imunologia , Macrófagos/imunologia , Plasmócitos/imunologia , Sepse/imunologia , Adenosina/metabolismo , Animais , Antígenos CD/metabolismo , Apirase/metabolismo , Reprogramação Celular/imunologia , Macrófagos/metabolismo , Camundongos , Plasmócitos/metabolismo , Receptor A2A de Adenosina/imunologia , Receptor A2A de Adenosina/metabolismo , Sepse/metabolismo
7.
Clin Transl Immunology ; 10(6): e1280, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34136216

RESUMO

OBJECTIVES: Inhibitors of bromodomain and extra terminal domain (BET) proteins are a new and growing class of anti-cancer drugs, which decrease oncogene expression by targeting superenhancers. Antibody production is another physiological process relying on superenhancers, and it remains to be clarified whether potential immunomodulatory properties of BET inhibitors might impact humoral immunity and allergy. METHODS: We thus evaluated humoral immune responses and their Th2 context in vitro and in vivo in mice following treatment with the classical BET-inhibitor JQ1. We quantified immunoglobulin (Ig) and antibody production by B cells either stimulated in vitro or obtained from immunised mice. JQ1 effects on class switching and activation-induced deaminase loading were determined, together with modifications of B, T follicular helper (Tfh) and T helper 2 (Th2) populations. JQ1 was finally tested in B-cell-dependent models of immune disorders. RESULTS: Bromodomain and extra terminal domain inhibition reduced class switching, Ig expression on B cells and antibody secretion and was correlated with decreased numbers of Tfh cells. However, JQ1 strongly increased the proportion of GATA3+ Th2 cells and the secretion of corresponding cytokines. In a mouse allergic model of lung inflammation, JQ1 did not affect eosinophil infiltration or mucus production but enhanced Th2 cytokine production and aggravated clinical manifestations. CONCLUSION: Altogether, BET inhibition thus interweaves intrinsic negative effects on B cells with a parallel complex reshaping of T-cell polarisation which can increase type 2 cytokines and eventually promote B-cell-dependent immunopathology. These opposite and potentially hazardous immunomodulatory effects raise concerns for clinical use of BET inhibitors in patients with immune disorders.

8.
Immunohorizons ; 5(5): 273-283, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33958388

RESUMO

Cystic fibrosis is associated with chronic Pseudomonas aeruginosa colonization and inflammation. The role of MyD88, the shared adapter protein of the proinflammatory TLR and IL-1R families, in chronic P. aeruginosa biofilm lung infection is unknown. We report that chronic lung infection with the clinical P. aeruginosa RP73 strain is associated with uncontrolled lung infection in complete MyD88-deficient mice with epithelial damage, inflammation, and rapid death. Then, we investigated whether alveolar or myeloid cells contribute to heightened sensitivity to infection. Using cell-specific, MyD88-deficient mice, we uncover that the MyD88 pathway in myeloid or alveolar epithelial cells is dispensable, suggesting that other cell types may control the high sensitivity of MyD88-deficient mice. By contrast, IL-1R1-deficient mice control chronic P. aeruginosa RP73 infection and IL-1ß Ab blockade did not reduce host resistance. Therefore, the IL-1R1/MyD88 pathway is not involved, but other IL-1R or TLR family members need to be investigated. Our data strongly suggest that IL-1 targeted neutralizing therapies used to treat inflammatory diseases in patients unlikely reduce host resistance to chronic P. aeruginosa infection.


Assuntos
Interleucina-1beta/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Receptores Tipo I de Interleucina-1/imunologia , Animais , Humanos , Imunidade Inata , Interleucina-1beta/genética , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Infecções por Pseudomonas/metabolismo , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais , Receptores Toll-Like/imunologia
9.
Kidney360 ; 2(11): 1793-1806, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-35372998

RESUMO

Background: Xanthinuria type II is a rare autosomal purine disorder. This recessive defect of purine metabolism remains an under-recognized disorder. Methods: Mice with targeted disruption of the molybdenum cofactor sulfurase (Mocos) gene were generated to enable an integrated understanding of purine disorders and evaluate pathophysiologic functions of this gene which is found in a large number of pathways and is known to be associated with autism. Results: Mocos-deficient mice die with 4 weeks of age due to renal failure of distinct obstructive nephropathy with xanthinuria, xanthine deposits, cystic tubular dilation, Tamm-Horsfall (uromodulin) protein (THP) deposits, tubular cell necrosis with neutrophils, and occasionally hydronephrosis with urolithiasis. Obstructive nephropathy is associated with moderate interstitial inflammatory and fibrotic responses, anemia, reduced detoxification systems, and important alterations of the metabolism of purines, amino acids, and phospholipids. Conversely, heterozygous mice expressing reduced MOCOS protein are healthy with no apparent pathology. Conclusions: Mocos-deficient mice develop a lethal obstructive nephropathy associated with profound metabolic changes. Studying MOCOS functions may provide important clues about the underlying pathogenesis of xanthinuria and other diseases requiring early diagnosis.


Assuntos
Nefropatias , Erros Inatos do Metabolismo da Purina-Pirimidina , Urolitíase , Animais , Nefropatias/genética , Camundongos , Erros Inatos do Metabolismo da Purina-Pirimidina/complicações , Urolitíase/genética , Xantina , Xantina Desidrogenase
10.
Cell Commun Signal ; 18(1): 141, 2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32894139

RESUMO

BACKGROUND: Low molecular weight carrageenan (Cg) is a seaweed-derived sulfated polysaccharide widely used as inflammatory stimulus in preclinical studies. However, the molecular mechanisms of Cg-induced inflammation are not fully elucidated. The present study aimed to investigate the molecular basis involved in Cg-induced macrophages activation and cytokines production. METHODS: Primary culture of mouse peritoneal macrophages were stimulated with Kappa Cg. The supernatant and cell lysate were used for ELISA, western blotting, immunofluorescence. Cg-induced mouse colitis was also developed. RESULTS: Here we show that Cg activates peritoneal macrophages to produce pro-inflammatory cytokines such as TNF and IL-1ß. While Cg-induced TNF production/secretion depends on TLR4/MyD88 signaling, the production of pro-IL-1ß relies on TLR4/TRIF/SYK/reactive oxygen species (ROS) signaling pathway. The maturation of pro-IL1ß into IL-1ß is dependent on canonical NLRP3 inflammasome activation via Pannexin-1/P2X7/K+ efflux signaling. In vivo, Cg-induced colitis was reduced in mice in the absence of NLRP3 inflammasome components. CONCLUSIONS: In conclusion, we unravel a critical role of the NLRP3 inflammasome in Cg-induced pro-inflammatory cytokines production and colitis, which is an important discovery on the pro-inflammatory properties of this sulfated polysaccharide for pre-clinical studies. Video abstract Carrageenan (Cg) is one the most used flogistic stimulus in preclinical studies. Nevertheless, the molecular basis of Cg-induced inflammation is not totally elucidated. Herein, Lopes et al. unraveled the molecular basis for Cg-induced macrophages production of biological active IL-1ß. The Cg-stimulated macrophages produces pro-IL-1ß depends on TLR4/TRIF/Syk/ROS, whereas its processing into mature IL-1ß is dependent on the canonical NLRP3 inflammasome.


Assuntos
Carragenina/imunologia , Citocinas/imunologia , Ativação de Macrófagos , Macrófagos Peritoneais/imunologia , Animais , Células Cultivadas , Inflamassomos/imunologia , Inflamação/imunologia , Interleucina-1beta/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Fator de Necrose Tumoral alfa/imunologia
11.
J Leukoc Biol ; 108(4): 1215-1223, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32745297

RESUMO

Macrophages are highly plastic cells, responding to diverse environmental stimuli to acquire different functional phenotypes. Signaling through MAPKs has been reported to regulate the differentiation of macrophages, but the role of ERK5 in IL-4-mediated M2 macrophage differentiation is still unclear. Here, we showed that the ERK5 signaling pathway plays a critical role in IL-4-induced M2 macrophage differentiation. Pharmacologic inhibition of MEK5, an upstream activator of ERK5, markedly reduced the expression of classical M2 markers, such as Arg-1, Ym-1, and Fizz-1, as well as the production of M2-related chemokines and cytokines, CCL22, CCL17, and IGF-1 in IL-4-stimulated macrophages. Moreover, pharmacologic inhibition of ERK5 also decreased the expression of several M2 markers induced by IL-4. In accordance, myeloid cell-specific Erk5 depletion (Erk5∆mye ), using LysMcre /Erk5f/f mice, confirmed the involvement of ERK5 in IL-4-induced M2 polarization. Mechanistically, the inhibition of ERK5 did not affect STAT3 or STAT6 phosphorylation, suggesting that ERK5 signaling regulates M2 differentiation in a STAT3 and STAT6-independent manner. However, genetic deficiency or pharmacologic inhibition of the MEK5/ERK5 pathway reduced the expression of c-Myc in IL-4-activated macrophages, which is a critical transcription factor involved in M2 differentiation. Our study thus suggests that the MEK5/ERK5 signaling pathway is crucial in IL-4-induced M2 macrophage differentiation through the induction of c-Myc expression.


Assuntos
Diferenciação Celular/imunologia , Interleucina-4/imunologia , MAP Quinase Quinase 5/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/imunologia , Proteína Quinase 7 Ativada por Mitógeno/imunologia , Proteínas Proto-Oncogênicas c-myc/imunologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Diferenciação Celular/genética , Regulação da Expressão Gênica/imunologia , Interleucina-4/genética , MAP Quinase Quinase 5/genética , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteínas Proto-Oncogênicas c-myc/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/imunologia
12.
Front Immunol ; 11: 1622, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849550

RESUMO

Cigarette smoke (CS) is the major cause of chronic lung injuries, such as chronic obstructive pulmonary disease (COPD). In patients with severe COPD, tertiary lymphoid follicles containing B lymphocytes and B cell-activating factor (BAFF) overexpression are associated with disease severity. In addition, BAFF promotes adaptive immunity in smokers and mice chronically exposed to CS. However, the role of BAFF in the early phase of innate immunity has never been investigated. We acutely exposed C57BL/6J mice to CS and show early BAFF expression in the bronchoalveolar space and lung tissue that correlates to airway neutrophil and macrophage influx. Immunostaining analysis revealed that neutrophils are the major source of BAFF. We confirmed in vitro that neutrophils secrete BAFF in response to cigarette smoke extract (CSE) stimulation. Antibody-mediated neutrophil depletion significantly dampens lung inflammation to CS exposure but only partially decreases BAFF expression in lung tissue and bronchoalveolar space suggesting additional sources of BAFF. Importantly, BAFF deficient mice displayed decreased airway neutrophil recruiting chemokines and neutrophil influx while the addition of exogenous BAFF significantly enhanced this CS-induced neutrophilic inflammation. This demonstrates that BAFF is a key proinflammatory cytokine and that innate immune cells in particular neutrophils, are an unconsidered source of BAFF in early stages of CS-induced innate immunity.


Assuntos
Fator Ativador de Células B/biossíntese , Exposição por Inalação/efeitos adversos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Pneumonia/etiologia , Pneumonia/metabolismo , Poluição por Fumaça de Tabaco/efeitos adversos , Animais , Fator Ativador de Células B/genética , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Infiltração de Neutrófilos , Pneumonia/patologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Fumar Tabaco/efeitos adversos
13.
Front Immunol ; 11: 144, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32161582

RESUMO

Airborne ozone exposure causes severe lung injury and inflammation. The aryl hydrocarbon Receptor (AhR) (1), activated in pollutant-induced inflammation, is critical for cytokine production, especially IL-22 and IL-17A. The role of AhR in ozone-induced lung inflammation is unknown. We report here that chronic ozone exposure activates AhR with increased tryptophan and lipoxin A4 production in mice. AhR-/- mice show increased lung inflammation, airway hyperresponsiveness, and tissue remodeling with an increased recruitment of IL-17A and IL-22-expressing cells in comparison to control mice. IL-17A- and IL-22-neutralizing antibodies attenuate lung inflammation in AhR-/- and control mice. Enhanced lung inflammation and recruitment of ILC3, ILC2, and T cells were observed after T cell-specific AhR depletion using the AhRCD4cre-deficient mice. Together, the data demonstrate that ozone exposure activates AhR, which controls lung inflammation, airway hyperresponsiveness, and tissue remodeling via the reduction of IL-22 expression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Interleucinas/metabolismo , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Ozônio/efeitos adversos , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linfócitos T CD4-Positivos/imunologia , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucinas/genética , Interleucinas/imunologia , Lipoxinas/metabolismo , Lesão Pulmonar/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia/tratamento farmacológico , Receptores de Hidrocarboneto Arílico/genética , Receptores de Interleucina-17/genética , Hipersensibilidade Respiratória/tratamento farmacológico , Triptofano/metabolismo , Interleucina 22
14.
Front Immunol ; 10: 2550, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31787969

RESUMO

Macrophages are essential cells of the innate immune response against microbial infections, and they have the ability to adapt under both pro- and anti-inflammatory conditions and develop different functions. A growing body of evidence regarding a novel macrophage subpopulation that expresses CD3 has recently emerged. Here, we explain that human circulating monocytes can be differentiated into CD3+TCRαß+ and CD3+TCRαß- macrophages. Both cell subpopulations express on their cell surface HLA family molecules, but only the CD3+TCRαß+ macrophage subpopulation co-express CD1 family molecules and transmembrane TNF (tmTNF). CD3+TCRαß+ macrophages secrete IL-1ß, IL-6 IP-10, and MCP-1 by both tmTNF- and CD3-dependent pathways, while CD3+TCRαß- macrophages specifically produce IFN-γ, TNF, and MIP-1ß by a CD3-dependent pathway. In this study, we also used a mouse model of BCG-induced pleurisy and demonstrated that CD3+ myeloid cells (TCRαß+ and TCRαß- cells) are increased at the infection sites during the acute phase (2 weeks post-infection). Interestingly, cell increment was mediated by tmTNF, and the soluble form of TNF was dispensable. BCG-infection also induced the expression of TNF receptor 2 on CD3+ myeloid cells, which increased after BCG-infection, suggesting that the tmTNF/TNFRs axis plays an important role in the presence or function of these cells in tuberculosis.


Assuntos
Complexo CD3/imunologia , Citocinas/metabolismo , Macrófagos/imunologia , Animais , Apresentação de Antígeno , Vacina BCG/administração & dosagem , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Leucócitos Mononucleares/imunologia , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Pleurisia/induzido quimicamente , Pleurisia/imunologia , Fator de Necrose Tumoral alfa/imunologia
15.
Sci Rep ; 9(1): 14848, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31619733

RESUMO

Cigarette smoke exposure is a leading cause of chronic obstructive pulmonary disease (COPD), a major health issue characterized by airway inflammation with fibrosis and emphysema. Here we demonstrate that acute exposure to cigarette smoke causes respiratory barrier damage with the release of self-dsDNA in mice. This triggers the DNA sensor cGAS (cyclic GMP-AMP synthase) and stimulator of interferon genes (STING), driving type I interferon (IFN I) dependent lung inflammation, which are attenuated in cGAS, STING or type I interferon receptor (IFNAR) deficient mice. Therefore, we demonstrate a critical role of self-dsDNA release and of the cGAS-STING-type I interferon pathway upon cigarette smoke-induced damage, which may lead to therapeutic targets in COPD.


Assuntos
DNA/metabolismo , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Pneumonia/metabolismo , Enfisema Pulmonar/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Poluição por Fumaça de Tabaco/efeitos adversos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sequências Repetitivas de Ácido Nucleico
16.
Front Immunol ; 10: 2169, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31608051

RESUMO

Ozone exposure causes irritation, airway hyperreactivity (AHR), inflammation of the airways, and destruction of alveoli (emphysema), the gas exchange area of the lung in human and mice. This review focuses on the acute disruption of the respiratory epithelial barrier in mice. A single high dose ozone exposure (1 ppm for 1 h) causes first a break of the bronchiolar epithelium within 2 h with leak of serum proteins in the broncho-alveolar space, disruption of epithelial tight junctions and cell death, which is followed at 6 h by ROS activation, AHR, myeloid cell recruitment, and remodeling. High ROS levels activate a novel PGAM5 phosphatase dependent cell-death pathway, called oxeiptosis. Bronchiolar cell wall damage and inflammation upon a single ozone exposure are reversible. However, chronic ozone exposure leads to progressive and irreversible loss of alveolar epithelial cells and alveoli with reduced gas exchange space known as emphysema. It is further associated with chronic inflammation and fibrosis of the lung, resembling other environmental pollutants and cigarette smoke in pathogenesis of asthma, and chronic obstructive pulmonary disease (COPD). Here, we review recent data on the mechanisms of ozone induced injury on the different cell types and pathways with a focus on the role of the IL-1 family cytokines and the related IL-33. The relation of chronic ozone exposure induced lung disease with asthma and COPD and the fact that ozone exacerbates asthma and COPD is emphasized.


Assuntos
Barreira Alveolocapilar/imunologia , Ozônio/toxicidade , Mucosa Respiratória/imunologia , Doença Aguda , Animais , Asma/induzido quimicamente , Asma/imunologia , Asma/patologia , Barreira Alveolocapilar/patologia , Fumar Cigarros/efeitos adversos , Fumar Cigarros/imunologia , Humanos , Camundongos , Fosfoproteínas Fosfatases/imunologia , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/patologia , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/patologia , Enfisema Pulmonar/induzido quimicamente , Enfisema Pulmonar/imunologia , Enfisema Pulmonar/patologia , Espécies Reativas de Oxigênio/imunologia , Mucosa Respiratória/patologia , Junções Íntimas/imunologia , Junções Íntimas/patologia
17.
Trends Immunol ; 40(8): 719-734, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31262653

RESUMO

Self-DNA sensing by the immune system has emerged as a key contributing response in the pathogenesis of cancer and autoimmune diseases. Recent studies have established that release of nuclear and mitochondrial DNA can also drive lung inflammatory diseases. Here, we review the latest advances on self-DNA sensing and signaling, the influence of these pathways on lung inflammation, and how these findings contribute to our understanding of basic mechanisms of innate immunity. Within a dozen DNA sensors, the cGAS/STING, inflammasomes and Toll-Like Receptor pathways are central to nucleic acid sensing. We propose a key role for the STING pathway in self-DNA sensing in inflammatory lung conditions, and identify major remaining questions that may further our understanding and potential to control self-DNA sensing and innate immune activation.


Assuntos
DNA/imunologia , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno/imunologia , Pneumonia/etiologia , Pneumonia/metabolismo , Animais , Autoimunidade , Biomarcadores , Suscetibilidade a Doenças/imunologia , Humanos , Imunidade Inata , Inflamassomos/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais
18.
Cell Rep ; 27(9): 2649-2664.e5, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31141689

RESUMO

Lung inflammation induced by silica impairs host control of tuberculosis, yet the underlying mechanism remains unclear. Here, we show that silica-driven exacerbation of M. tuberculosis infection associates with raised type 2 immunity. Silica increases pulmonary Th2 cell and M2 macrophage responses, while reducing type 1 immunity after M. tuberculosis infection. Silica induces lung damage that prompts extracellular self-DNA release and activates STING. This STING priming potentiates M. tuberculosis DNA sensing by and activation of cGAS/STING, which triggers enhanced type I interferon (IFNI) response and type 2 immunity. cGAS-, STING-, and IFNAR-deficient mice are resistant to silica-induced exacerbation of M. tuberculosis infection. Thus, silica-induced self-DNA primes the host response to M. tuberculosis-derived nucleic acids, which increases type 2 immunity while reducing type 1 immunity, crucial for controlling M. tuberculosis infection. These data show how cGAS/STING pathway activation, at the crossroads of sterile inflammation and infection, may affect the host response to pathogens such as M. tuberculosis.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Proteínas de Membrana/fisiologia , Mycobacterium tuberculosis/imunologia , Pneumonia/complicações , Dióxido de Silício/toxicidade , Tuberculose/etiologia , Animais , Células Dendríticas , Fator Regulador 3 de Interferon/fisiologia , Interferon Tipo I/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nucleotidiltransferases/fisiologia , Pneumonia/induzido quimicamente , Receptor de Interferon alfa e beta/fisiologia , Transdução de Sinais , Tuberculose/metabolismo , Tuberculose/patologia
19.
Sci Rep ; 9(1): 5297, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30923339

RESUMO

TNF plays a critical role in mononuclear cell recruitment during acute Bacillus Calmette-Guérin (BCG) infection leading to an effective immune response with granuloma formation, but may also cause tissue injury mediated by TNFR1 or TNFR2. Here we investigated the role of myeloid and T cell specific TNFR1 and R2 expression, and show that absence of TNFR1 in myeloid cells attenuated liver granuloma formation and liver injury in response to acute BCG infection, while TNFR2 expressed in myeloid cells contributed only to liver injury. TNFR1 was the main receptor controlling cytokine production by liver mononuclear cells after antigenic specific response, modified CD4/CD8 ratio and NK, NKT and regulatory T cell recruitment. Further analysis of CD11b+CD3+ phagocytic cells revealed a TCRαß expressing subpopulation of unknown function, which increased in response to BCG infection dependent of TNFR1 expression on myeloid cells. In conclusion, TNFR1 expressed by myeloid cells plays a critical role in mononuclear cell recruitment and injury of the liver after BCG infection.


Assuntos
Vacina BCG/efeitos adversos , Granuloma/imunologia , Hepatite/imunologia , Mycobacterium bovis/patogenicidade , Células Mieloides/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Vacina BCG/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Granuloma/microbiologia , Granuloma/patologia , Hepatite/microbiologia , Hepatite/patologia , Humanos , Fígado/citologia , Fígado/imunologia , Fígado/patologia , Camundongos , Camundongos Knockout , Mycobacterium bovis/imunologia , Células Mieloides/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Vacinas Vivas não Atenuadas/efeitos adversos
20.
Nat Commun ; 9(1): 5226, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30523277

RESUMO

Silica particles induce lung inflammation and fibrosis. Here we show that stimulator of interferon genes (STING) is essential for silica-induced lung inflammation. In mice, silica induces lung cell death and self-dsDNA release in the bronchoalveolar space that activates STING pathway. Degradation of extracellular self-dsDNA by DNase I inhibits silica-induced STING activation and the downstream type I IFN response. Patients with silicosis have increased circulating dsDNA and CXCL10 in sputum, and patients with fibrotic interstitial lung disease display STING activation and CXCL10 in the lung. In vitro, while mitochondrial dsDNA is sensed by cGAS-STING in dendritic cells, in macrophages extracellular dsDNA activates STING independent of cGAS after silica exposure. These results reveal an essential function of STING-mediated self-dsDNA sensing after silica exposure, and identify DNase I as a potential therapy for silica-induced lung inflammation.


Assuntos
DNA/metabolismo , Proteínas de Membrana/metabolismo , Pneumonia/metabolismo , Dióxido de Silício/metabolismo , Animais , Células Cultivadas , Quimiocina CXCL10/metabolismo , DNA/genética , Células Dendríticas/metabolismo , Humanos , Macrófagos/metabolismo , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia/genética , Dióxido de Silício/química , Silicose/metabolismo , Escarro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA