Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
1.
Environ Sci Pollut Res Int ; 29(17): 24445-24456, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35064883

RESUMO

Fine particulate matter (PM2.5) has been the primary air pollutant and the fourth leading risk factor for disease and death in the world. Exposure to PM2.5 is related to activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, but the mechanism of PM2.5 affecting the NLRP3 inflammasome is still unclear. Previous studies have shown that PM2.5 can cause alterations in adenosine triphosphate (ATP), and an increase in extracellular ATP and a decrease in intracellular ATP can trigger the activation process of the NLRP3 inflammasome. Therefore, we emphasize that ATP changes may be the central link and key mechanism of PM2.5 exposure that activates the NLRP3 inflammasome. This review briefly elucidates and summarizes how PM2.5 acts on ATP and subsequently further impacts the NLRP3 inflammasome. Investigation of ATP changes due to exposure to PM2.5 may be essential to regulate NLRP3 inflammasome activation and treat inflammation-related diseases such as coronavirus disease 2019 (COVID-19).


Assuntos
COVID-19 , Inflamassomos , Trifosfato de Adenosina , Humanos , Inflamassomos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Material Particulado
2.
Front Immunol ; 12: 746032, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659244

RESUMO

Aberrant inflammasome activation contributes to various chronic inflammatory diseases; however, pyroptosis of inflammasome-active cells promptly terminates local inflammasome response. Molecular mechanisms underlying prolonged inflammasome signaling thus require further elucidation. Here, we report that neutrophil-specific resistance to pyroptosis and NLRP3 desensitization can facilitate sustained inflammasome response and interleukin-1ß secretion. Unlike macrophages, inflammasome-activated neutrophils did not undergo pyroptosis, indicated by using in vitro cell-based assay and in vivo mouse model. Intriguingly, danger-associated molecular patterns (DAMP)-rich milieu in the inflammatory region significantly abrogated NLRP3-activating potential of macrophages, but not of neutrophils. This macrophage-specific NLRP3 desensitization was associated with DAMP-induced mitochondrial depolarization that was not observed in neutrophils due to a lack of SARM1 expression. Indeed, valinomycin-induced compulsory mitochondrial depolarization in neutrophils restored inflammasome-dependent cell death and ATP-induced NLRP3 desensitization in neutrophils. Alongside prolonged inflammasome-activating potential, neutrophils predominantly secreted interleukin-1ß rather than other proinflammatory cytokines upon NLRP3 stimulation. Furthermore, inflammasome-activated neutrophils did not trigger efferocytosis-mediated M2 macrophage polarization essential for the initiation of inflammation resolution. Taken together, our results indicate that neutrophils can prolong inflammasome response via mitochondria-dependent resistance to NLRP3 desensitization and function as major interleukin-1ß-secreting cells in DAMP-rich inflammatory region.


Assuntos
Alarminas/análise , Inflamassomos/fisiologia , Inflamação/imunologia , Neutrófilos/imunologia , Animais , Proteínas do Domínio Armadillo/fisiologia , Citocinas/biossíntese , Proteínas do Citoesqueleto/fisiologia , Feminino , Interleucina-1beta/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Neutrófilos/efeitos dos fármacos , Fagocitose , Proteínas de Ligação a Fosfato/metabolismo , Piroptose , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/metabolismo , Organismos Livres de Patógenos Específicos
3.
Cells ; 10(9)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34572082

RESUMO

Aortic diseases comprise aneurysms, dissections, and several other pathologies. In general, aging is associated with a slow but progressive dilation of the aorta, along with increased stiffness and pulse pressure. The progression of aortic disease is characterized by subclinical development or acute presentation. Recent evidence suggests that inflammation participates causally in different clinical manifestations of aortic diseases. As of yet, diagnostic imaging and surveillance is mainly based on ultrasonography, computed tomography (CT), and magnetic resonance imaging (MRI). Little medical therapy is available so far to prevent or treat the majority of aortic diseases. Endovascular therapy by the introduction of covered stentgrafts provides the main treatment option, although open surgery and implantation of synthetic grafts remain necessary in many situations. Because of the risks associated with surgery, there is a need for identification of pharmaceutical targets interfering with the pathophysiology of aortic remodeling. The participation of innate immunity and inflammasome activation in different cell types is common in aortic diseases. This review will thus focus on inflammasome activities in vascular cells of different chronic and acute aortic diseases and discuss their role in development and progression. We will also identify research gaps and suggest promising therapeutic targets, which may be used for future medical interventions.


Assuntos
Aorta , Doenças da Aorta , Inflamassomos/metabolismo , Aorta/citologia , Aorta/patologia , Aorta/fisiologia , Aneurisma Aórtico/metabolismo , Aneurisma Aórtico/fisiopatologia , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/fisiopatologia , Doenças da Aorta/metabolismo , Doenças da Aorta/fisiopatologia , Proteínas de Ligação a DNA/metabolismo , Sistemas de Liberação de Medicamentos , Células Endoteliais/metabolismo , Humanos , Imuno-Histoquímica , Inflamassomos/fisiologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Interleucina-1beta/metabolismo , Linfócitos/metabolismo , Macrófagos/metabolismo , Miócitos de Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
4.
Front Immunol ; 12: 710110, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34421915

RESUMO

Inflammasomes are fundamental innate immune mechanisms that promote inflammation and induce an inflammatory form of programmed cell death, pyroptosis. Pyroptotic inflammasome has been reported to be closely associated with tumorigenesis and prognosis of multiple cancers. Emerging studies show that the inflammasome assembly into a higher-order supramolecular complex has been utilized to evaluate the status of the innate immune response. The inflammasomes are now regarded as cellular signaling hubs of the innate immunity that drive the production of inflammatory cytokines and consequent recruitment of immune cells to the tumor sites. Herein, we provided an overview of molecular characteristics and biological properties of canonical and non-canonical inflammasome signaling in cancer immunology and immunotherapy. We also focus on the mechanism of regulating pyroptotic inflammasome in tumor cells, as well as the potential roles of inflammasome-mediated pyroptotic cell death in cancers, to explore the potential diagnostic and therapeutic markers contributing to the prevention and treatment of cancers.


Assuntos
Inflamassomos/fisiologia , Neoplasias/imunologia , Carcinogênese , Armadilhas Extracelulares/fisiologia , Humanos , Imunidade Inata , Imunoterapia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias/terapia , Transdução de Sinais/fisiologia , Microambiente Tumoral
5.
Front Immunol ; 12: 643149, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34177893

RESUMO

Fibrosis is the final common pathway of inflammatory diseases in various organs. The inflammasomes play an important role in the progression of fibrosis as innate immune receptors. There are four main members of the inflammasomes, such as NOD-like receptor protein 1 (NLRP1), NOD-like receptor protein 3 (NLRP3), NOD-like receptor C4 (NLRC4), and absent in melanoma 2 (AIM2), among which NLRP3 inflammasome is the most studied. NLRP3 inflammasome is typically composed of NLRP3, ASC and pro-caspase-1. The activation of inflammasome involves both "classical" and "non-classical" pathways and the former pathway is better understood. The "classical" activation pathway of inflammasome is that the backbone protein is activated by endogenous/exogenous stimulation, leading to inflammasome assembly. After the formation of "classic" inflammasome, pro-caspase-1 could self-activate. Caspase-1 cleaves cytokine precursors into mature cytokines, which are secreted extracellularly. At present, the "non-classical" activation pathway of inflammasome has not formed a unified model for activation process. This article reviews the role of NLRP1, NLRP3, NLRC4, AIM2 inflammasome, Caspase-1, IL-1ß, IL-18 and IL-33 in the fibrogenesis.


Assuntos
Fibrose/etiologia , Inflamassomos/fisiologia , Animais , Proteínas Adaptadoras de Sinalização CARD/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Caspase 1/fisiologia , Humanos , Inflamassomos/classificação , Interleucina-1beta/fisiologia , Interleucina-33/fisiologia , Rim/patologia , Cirrose Hepática/etiologia , Miocárdio/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas NLR/fisiologia , Fibrose Pulmonar/etiologia
6.
Front Immunol ; 12: 676088, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122438

RESUMO

As a highly inflammatory form of programmed cell death, pyroptosis is triggered by pro-inflammatory signals and associated with inflammation. It is characterized by cell swelling and large bubbles emerging from the plasma membrane, which release cytokines during inflammation. Compared with other types of cell death, pyroptosis has a distinct morphology and mechanism and involves special inflammasome cascade pathways. However, the inflammasome mechanism through which endometrial epithelial cell pyroptosis occurs in LPS-mediated inflammation remains unclear. We confirmed that there was an increased mRNA and protein expression of the IL-6, TNF-α, IL-1ß, IL-18 cytokines, the inflammasome molecules NLRP3, CASPASE-1, CASPASE-4, and GSDMD in LPS-induced primary bovine endometrial epithelial cells (BEECs) in an in vitro established inflammatory model using ELISA, real-time PCR (RT-PCR), vector construction and transfection, and Western blotting. Scanning electron microscopy and lactate dehydrogenase (LDH) activity assays revealed induced cell membrane rupture, which is the main characteristic of pyroptosis. In conclusion, the cytolytic substrate GSDMD's cleavage by caspase-1 or caspase-4 through the NLRP3 classical and non-classical inflammasome pathways, GSDMD N-terminus bind to the plasma membrane to form pores and release IL -18, IL-1ß cause cell death during LPS induced BEECs inflammation.


Assuntos
Endométrio/efeitos dos fármacos , Inflamassomos/fisiologia , Lipopolissacarídeos/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Piroptose/efeitos dos fármacos , Animais , Bovinos , Células Cultivadas , Citocinas/fisiologia , Endométrio/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Inflamação/etiologia , Transdução de Sinais/fisiologia
7.
Exp Cell Res ; 405(1): 112635, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34051241

RESUMO

Exosomes have been shown to have therapeutic potential for cerebral ischemic diseases. In this study, we investigated the neuroprotective effects of normoxic and hypoxic bone marrow mesenchymal stromal cells-derived exosomes (N-BM-MSCs-Exo and H-BM-MSCs-Exo, respectively) on oxygen-glucose deprivation (OGD) injury in mouse neuroblastoma N2a cells and rat primary cortical neurons. The proportions of dead cells in N2a and primary cortical neurons after OGD injury were significantly increased, and N-BM-MSCs-Exo (40 µg/ml) could reduce the ratios, noteworthily, the protective effects of H-BM-MSCs-Exo (40 µg/ml) were more potent. Western blotting analysis indicated that N-BM-MSCs-Exo decreased the expression of NLRP3, ASC, Caspase-1, GSDMD-N, cleaved IL-1ß and IL-18 in N2a cells. However, H-BM-MSCs-Exo (40 µg/ml) was more powerful in inhibiting the expression of these proteins in comparison with N-BM-MSCs-Exo. Similar results were obtained in primary cortical neurons. Immunofluorescence assays showed that after N-BM-MSCs-Exo and H-BM-MSCs-Exo treatment, the co-localization of NLRP3, ASC, Caspase-1 and the GSDMD translocation from the nucleus to the cytoplasm and membrane after OGD injury were reduced in N2a cells and primary cortical neurons, and H-BM-MSCs-Exo had a more obvious effect. In addition, N-BM-MSCs-Exo and H-BM-MSCs-Exo significantly reduced lactate dehydrogenase (LDH) release and the IL-18 levels in cell culture medium in N2a cells and primary cortical neurons. Once again H-BM-MSCs-Exo induced these effects more potently than N-BM-MSCs-Exo. All of these results demonstrated that N-BM-MSCs-Exo and H-BM-MSCs-Exo have significant neuroprotective effects against NLRP3 inflammasome-mediated pyroptosis. H-BM-MSCs-Exo has a more pronounced protective effect than N-BM-MSCs-Exo and may be used to ameliorate the progression of cerebral ischemia and hypoxia injury in patients.


Assuntos
Exossomos/fisiologia , Hipóxia/fisiopatologia , Células-Tronco Mesenquimais/citologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neuroblastoma/prevenção & controle , Neurônios/citologia , Piroptose , Animais , Córtex Cerebral/citologia , Córtex Cerebral/imunologia , Córtex Cerebral/lesões , Córtex Cerebral/metabolismo , Glucose/deficiência , Inflamassomos/fisiologia , Masculino , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Neuroblastoma/etiologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Transdução de Sinais
8.
Mediators Inflamm ; 2021: 1610251, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33967625

RESUMO

OBJECTIVE: Inflammatory bowel disease (IBD) is a chronic nonspecific inflammatory bowel disease with an unclear etiology. The active ingredients of traditional Chinese medicines (TCMs) exert anti-inflammatory, antitumor, and immunomodulatory effects, and their multitarget characteristics provide them with a unique advantage for treating IBD. However, the therapeutic effects and underlying mechanisms of Xi Lei San in treatment of IBD remain unknown. This study was designed to investigate whether Xi Lei San exerted an anti-inflammatory effect in IBD via a mechanism involving NLRP3 inflammasomes and autophagy. METHODS: We successfully established a rat model of dextran sulfate sodium- (DSS-) induced colitis as well as a cellular model of TNF-α-induced colitis. Xi Lei San and indirubin were identified by HPLC analysis. Rats were treated with Xi Lei San or alum crystals, and their body weights and morphology of intestinal tissues were examined. A western blot analysis was performed to determine the expression levels of inflammasome-related proteins and autophagy-related proteins, ELISA was performed to analyze IL-1ß, IL-18, and IL-33 concentrations, and flow cytometry was used to monitor cell apoptosis and ROS levels. RESULTS: Xi Lei San and indirubin were identified by HPLC analysis. We found that Xi Lei San could significantly increase the weights of rats and improve the structure of the intestinal tissues in DSS-induced colitis model rats. We also found that Xi Lei San significantly inhibited NLRP3 inflammasome activity, reduced the levels of inflammatory cytokines, and suppressed autophagy in DSS-induced colitis model rats. In vitro experiments revealed that Xi Lei San could repress apoptosis as well as ROS and inflammatory cytokine production in TNF-α-induced CACO2 cells by reducing the activity of NLRP3 inflammasomes and autophagy. CONCLUSIONS: Our findings showed that Xi Lei San significantly ameliorated IBD by inhibiting NLRP3 inflammasome, autophagy, and oxidative stress.


Assuntos
Autofagia/fisiologia , Inflamassomos/fisiologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Medicina Tradicional Chinesa , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células CACO-2 , Citocinas/biossíntese , Sulfato de Dextrana , Feminino , Humanos , Indóis/farmacologia , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/imunologia , Ratos , Ratos Sprague-Dawley
9.
Front Immunol ; 12: 642855, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33968032

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a relentlessly progressive disease harboring significant morbidity and mortality despite recent advances in therapy. Regardless of disease severity acute exacerbations (IPF-AEs) may occur leading to considerable loss of function and are the leading cause of death in IPF. Histologic features of IPF-AE are very similar to acute respiratory distress syndrome (ARDS), but the underlying mechanisms are incompletely understood. We investigated the role of the NLRP3 inflammasome in IPF and IPF-AE. Bronchoalveolar lavage (BAL) cells were sampled from patients with IPF (n = 32), IPF-AE (n = 10), ARDS (n = 7) and healthy volunteers (HV, n = 37) and the NLRP3-inflammasome was stimulated in-vitro. We found the NLRP3 inflammasome to be hyper-inducible in IPF compared to HV with increased IL-1ß and pro-IL-1ß levels on ELISA upon stimulation as well as increased caspase-1 activity measured by caspase-1p20 immunoblotting. In IPF-AE, IL-1ß was massively elevated to an extent similar to ARDS. To evaluate potential mechanisms, we co-cultured BAL cells with radiated A549 cells (a model to simulate apoptotic alveolar epithelial cells), which led to increased NLRP3 mRNA expression and increased caspase-1 dependent IL-1ß production. In the presence of a reactive oxygen species (ROS) inhibitor (diphenyleneiodonium) and a cathepsin B inhibitor (E64D), NLRP3 expression was suppressed indicating that induction of NLRP3 activation following efferocytosis of apoptotic A549 cells is mediated via ROS and cathepsin-B. In summary, we present evidence of involvement of the NLRP3 inflammasome-caspase pathway in the pathogenesis of IPF-AE, similarly to ARDS, which may be mediated by efferocytosis of apoptotic alveolar epithelial cells in IPF.


Assuntos
Apoptose , Caspase 1/fisiologia , Fibrose Pulmonar Idiopática/complicações , Inflamassomos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Células A549 , Doença Aguda , Adulto , Idoso , Idoso de 80 Anos ou mais , Catepsina B/fisiologia , Feminino , Humanos , Interleucina-1/fisiologia , Masculino , Pessoa de Meia-Idade , Precursores de Proteínas/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Adulto Jovem
10.
Life Sci ; 277: 119489, 2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-33862118

RESUMO

AIMS: Nod-like receptor family pyrin domain containing 3 (NLRP3) may play an important role in neuropathic pain. Treatment for trigeminal neuropathic pain remains a challenge, as common drugs either do not demonstrate beneficial therapeutic effects or induce intolerance in patients. MAIN METHODS: In a rat model of trigeminal neuropathic pain, pain caused by the malpositioning of dental implants is similar to that experienced by humans. We used masculine Sprague-Dawley rats with inferior alveolar nerve damage as a model to investigate the differential regulation of NLRP3. First, we confirmed the level of NLRP3 in the medullary dorsal horn and variation of pain response behavior after silencing the expression of NLRP3 inflammasome bodies in rats with trigeminal neuropathic pain. Second, under localized anesthesia, we extracted the lower left second molar, implanted a micro-dental implant, and deliberately injured the inferior alveolar nerve. KEY FINDINGS: After nerve damage, the level of NLRP3-related inflammasomes was upregulated in microglia and the expression of a component of the inflammasome gradually increased during postoperative days 3-21. The suppression of adenovirus-shRNA-NLRP3 on postoperative day 1 markedly inhibited the expression of pro-inflammatory cytokines and the activation of the inflammasome and mechanical allodynia. Furthermore, it attenuated cell death in microglia, as evidenced by increased Bcl-2, Bcl-xL, Bax, and Bik expression. SIGNIFICANCE: The level of NLRP3 in the dorsal horn is a pivotal factor in trigeminal neuropathic pain, and inhibition of the early expression of NLRP3 might serve as a potential therapeutic approach.


Assuntos
Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doenças do Nervo Trigêmeo/metabolismo , Animais , Encéfalo/metabolismo , Citocinas/metabolismo , Hiperalgesia/tratamento farmacológico , Inflamassomos/metabolismo , Inflamassomos/fisiologia , Masculino , Bulbo/metabolismo , Microglia/metabolismo , Neuralgia/tratamento farmacológico , Células do Corno Posterior/metabolismo , Ratos , Ratos Sprague-Dawley , Doenças do Nervo Trigêmeo/fisiopatologia , Neuralgia do Trigêmeo/tratamento farmacológico
11.
J Invest Dermatol ; 141(12): 2885-2894.e5, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33745908

RESUMO

Rosacea is a chronic inflammatory skin disease characterized by immune response-dependent erythema and pustules. Although the precise etiology of rosacea remains elusive, its pathogenesis is reportedly associated with an increased level of antimicrobial peptide LL-37. However, molecular mechanisms underlying the progression of rosacea via LL-37 remain poorly understood. Here, we examined the potential role of LL-37 in rosacea-like skin inflammatory phenotypes at a molecular level. Our in vitro data demonstrated that LL-37 promotes NLRP3-mediated inflammasome activation in lipopolysaccharide-primed macrophages, indicated by the processing of caspase-1 and IL-1ß. LL-37 was internalized into the cytoplasm of macrophages through P2X7 receptor-mediated endocytosis. Intracellular LL-37 triggered the assembly and activation of NLRP3-ASC inflammasome complex by facilitating lysosomal destabilization. Consistent with these in vitro results, intradermal LL-37 administration induced in vivo caspase-1 activation and ASC speck formation in the skin of Nlrp3-expressing, but not in Nlrp3-deficient, mice. Intradermal injection of LL-37 elicited profound recruitment of inflammatory Gr1+ cells and subsequent skin inflammation. However, LL-37-induced rosacea-like skin inflammation was significantly abrogated in Nlrp3-deficient mice. Furthermore, an NLRP3-specific inhibitor, MCC950, markedly reduced LL-37-triggered rosacea-like phenotypes. Taken together, our findings clearly indicate that NLRP3 inflammasome activation plays a crucial role in LL-37-induced skin inflammation and rosacea pathogenesis.


Assuntos
Peptídeos Catiônicos Antimicrobianos/efeitos adversos , Inflamassomos/fisiologia , Inflamação/induzido quimicamente , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Rosácea/induzido quimicamente , Animais , Caspase 1/metabolismo , Células Cultivadas , Feminino , Furanos/farmacologia , Indenos/farmacologia , Interleucina-1beta/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Sulfonamidas/farmacologia , Catelicidinas
12.
Theranostics ; 11(5): 2424-2441, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33500734

RESUMO

Rationale: Stimulation of the NLRP3 inflammasome by metabolic byproducts is known to result in inflammatory responses and metabolic diseases. However, how the host controls aberrant NLRP3 inflammasome activation remains unclear. PPARγ, a known regulator of energy metabolism, plays an anti-inflammatory role through the inhibition of NF-κB activation and additionally attenuates NLRP3-dependent IL-1ß and IL-18 production. Therefore, we hypothesized that PPARγ serves as an endogenous modulator that attenuates NLRP3 inflammasome activation in macrophages. Methods: Mouse peritoneal macrophages with exposure to a PPARγ agonist at different stages and the NLRP3 inflammasome-reconstituted system in HEK293T cells were used to investigate the additional anti-inflammatory effect of PPARγ on NLRP3 inflammasome regulation. Circulating mononuclear cells of obese patients with weight-loss surgery were used to identify the in vivo correlation between PPARγ and the NLRP3 inflammasome. Results: Exposure to the PPARγ agonist, rosiglitazone, during the second signal of NLRP3 inflammasome activation attenuated caspase-1 and IL-1ß maturation. Moreover, PPARγ interfered with NLRP3 inflammasome formation by decreasing NLRP3-ASC and NLRP3-NLRP3 interactions as well as NLRP3-dependent ASC oligomerization, which is mediated through interaction between the PPARγ DNA-binding domain and the nucleotide-binding and leucine-rich repeat domains of NLRP3. Furthermore, PPARγ was required to limit metabolic damage-associated molecular pattern-induced NLRP3 inflammasome activation in mouse macrophages. Finally, the mature caspase-1/PPARγ ratio was reduced in circulating mononuclear cells of obese patients after weight-loss surgery, which we define as an "NLRP3 accelerating index". Conclusions: These results revealed an additional anti-inflammatory role for PPARγ in suppressing NLRP3 inflammasome activation through interaction with NLRP3. Thus, our study highlights that PPARγ agonism may be a therapeutic option for targeting NLRP3-related metabolic diseases.


Assuntos
Inflamassomos/fisiologia , Inflamação/patologia , Macrófagos Peritoneais/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Obesidade/fisiopatologia , PPAR gama/agonistas , Rosiglitazona/farmacologia , Animais , Humanos , Hipoglicemiantes/farmacologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
13.
Life Sci ; 267: 118918, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33352170

RESUMO

The NLRP3 inflammasome regulates innate immune and inflammatory responses by promoting pro-inflammatory cytokines such as IL-18 and IL-1ß. NLRP3 is one of the main factors restricting the activation of the inflammasome, which is closely related to the abundance and localization of NLRP3. A substantial number of studies have focused on specifically targeting NLRP3 to develop inhibitors against NLRP3 inflammasome. Here, we succinctly review the regulation of NLRP3 expression at DNA/chromosome, transcriptional, post-transcriptional, and translation levels. These are critical for the fine regulation of the NLRP3 inflammasome.


Assuntos
Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Animais , Citocinas/metabolismo , Humanos , Imunidade Inata/imunologia , Inflamassomos/metabolismo , Inflamassomos/fisiologia , Inflamação/imunologia , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transdução de Sinais
14.
Shock ; 55(2): 274-282, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32769820

RESUMO

ABSTRACT: Neonatal sepsis leads to significant morbidity and mortality with the highest risk of death occurring in preterm (<37 weeks) and low birth weight (<2,500 g) infants. The neonatal immune system is developmentally immature with well-described defects in innate and adaptive immune responses. Immune adjuvants used to enhance the vaccine response have emerged as potential therapeutic options, stimulating non-specific immunity and preventing sepsis mortality. Aluminum salts ("alum") have been used as immune adjuvants for over a century, but their mechanism of action remains poorly understood. This study aims to identify potential mechanisms by which pretreatment with alum induces host protective immunity to polymicrobial sepsis in neonatal mice. Utilizing genetic and cell-depletion studies, we demonstrate here that the prophylactic administration of aluminum adjuvants in neonatal mice improves sepsis survival via activation of the nucleotide oligomerization domain-like receptor family, pyrin-domain-containing 3 inflammasome and dendritic cell activation. Furthermore, this beneficial effect is dependent on myeloid, non-granulocytic Gr1-positive cells, and MyD88-signaling pathway activation. These findings suggest a promising therapeutic role for aluminum-based vaccine adjuvants to prevent development of neonatal sepsis and improve mortality in this highly vulnerable population.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Compostos de Alúmen/uso terapêutico , Inflamassomos/fisiologia , Células Mieloides/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Sepse Neonatal/tratamento farmacológico , Sepse Neonatal/mortalidade , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Granulócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Taxa de Sobrevida
15.
Neuromolecular Med ; 22(4): 474-483, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33073305

RESUMO

The CD137L-CD137 axis is a potent co-stimulatory immune checkpoint regulator that forms a bidirectional signaling pathway between the CD137 ligand (CD137L) and CD137 receptor to regulate immunological activities. This study investigated the potential involvement of the CD137L-CD137 axis on inflammasome-associated brain injury and neurological deficits in a mouse model of focal ischemic stroke. Cerebral ischemia was induced in male C57BL/6J wild-type (WT), CD137L-deficient (CD137L KO) and CD137-deficient (CD137 KO) mice by middle cerebral artery occlusion (MCAO; 60 min), followed by reperfusion (6 h and 24 h). Brain infarct volume and neurological deficit scores were significantly lower in both CD137L KO and CD137 KO mice compared to WT controls. Moreover, CD137L-deficient brains had significantly lower levels of the pyroptotic protein, NT-Gasdermin D, while CD137-deficient brains had significantly lower levels of the pro-apoptotic proteins, cleaved caspase-3, pyroptotic protein, NT-Gasdermin D, and of the secondary pyroptotic protein NT-Gasdermin E, following ischemic stroke. This protection by CD137L and CD137 deletion was associated with a significant decrease in inflammasome signaling. In conclusion, our data provide evidence for the first time that the CD137L-CD137 axis contributes to brain injury and neurological deficits by activating the inflammasome signaling pathway following ischemic stroke.


Assuntos
Ligante 4-1BB/fisiologia , Infarto da Artéria Cerebral Média/metabolismo , Inflamassomos/fisiologia , AVC Isquêmico/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/fisiologia , Ligante 4-1BB/deficiência , Alarminas/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Dano Encefálico Crônico/etiologia , Infarto Cerebral/etiologia , Infarto Cerebral/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , AVC Isquêmico/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Ligação a Fosfato/metabolismo , Receptores de Estrogênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/fisiologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/deficiência
16.
Cell Death Dis ; 11(10): 901, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-33093455

RESUMO

Activation of the nucleotide-binding domain leucine-rich repeat and pyrin domain containing receptor 3 (NLRP3) inflammasome plays an important role in ocular neovascularization. In our study, we found that the expression and activation levels of NLRP3 inflammasome components, including NLRP3, an apoptosis-associated speck-like protein (ASC) containing caspase activation and recruitment domain (CARD) and caspase-1 (CAS1), were significantly upregulated. In addition, we found interleukin (IL)-1ß activity increased while IL-18 activity decreased in the retinas of oxygen-induced ischemic retinopathy (OIR) mice. MCC950, an inhibitor of NLRP3, reversed the IL-1ß/IL-18 activation pattern, inhibited the formation of retinal neovascularization (RNV), decreased the number of acellular capillaries and reduced leakage of retinal vessels. Moreover, MCC950 could regulate the expression of endothelial cell- and pericyte function-associated molecules, such as vascular endothelial growth factor (VEGF), VEGF receptor (VEGFR)1, VEGFR2, matrix metalloproteinase (MMP)2, MMP9, tissue inhibitor of metalloproteinases (TIMP)1, TIMP2, platelet-derived growth factor receptor-ß (PDGFR-ß), platelet-derived growth factor-B (PDGF-B), and angiopoietin2 (Ang2). In vitro, recombinant human (r)IL-18 and rIL-1ß regulated the expression of endothelial cell- and pericyte function-associated molecules and the proliferation and migration of endothelial cells and pericytes. We therefore determined that inhibiting the NLRP3 inflammasome with MCC950 can regulate the function of endothelial cells and pericytes by reversing the IL-1ß/IL-18 activation pattern to ameliorate RNV and leakage; thereby opening new avenues to treat RNV-associated ocular diseases.


Assuntos
Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Inflamassomos/fisiologia , Interleucina-18/fisiologia , Interleucina-1beta/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Sulfonas/farmacologia , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Furanos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Indenos , Isquemia/induzido quimicamente , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Recombinantes , Doenças Retinianas/induzido quimicamente , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Sulfonamidas , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Biochem Biophys Res Commun ; 533(4): 1184-1190, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33041005

RESUMO

The nucleotide-binding domain and leucine-rich repeat-containing family pyrin domain containing 3 (NLRP3) inflammasome is involved in various acute and chronic liver diseases, however, it is not clear whether NLRP3 contributes to d-Galactosamine (D-GalN) plus lipopolysaccharide (LPS)-induced acute liver failure (ALF). This study aims to investigate the role of NLRP3 inflammasome in D-GalN/LPS-induced fatal hepatitis. We found that Nlrp3-/- and WT mice showed similar mortality against a lethal dose of D-GalN/LPS treatment. Serum ALT and AST levels, as well as liver necrosis area and hepatocyte apoptosis, were not significantly different between Nlrp3-/- and WT mice at 6 h after D-GalN/LPS injection. Moreover, the numbers of intrahepatic F4/80+ cells and Ly6G+ cells were comparable in two genotype mice following D-GalN/LPS treatment. Besides, Nlrp3-/- mice had reduced IL-1ß levels but similar TNF-α, IL-6, and MCP-1 levels compared with WT mice upon D-GalN/LPS administration. Our findings revealed that NLRP3 ablation does not protect mice from D-GalN/LPS-induced fatal hepatitis and has a marginal effect on intrahepatic inflammatory response upon D-GalN/LPS treatment. This suggests that NLRP3 inflammasome does not appear to be a major contributor to D-GalN/LPS-induced ALF.


Assuntos
Falência Hepática Aguda/etiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Galactosamina , Inflamassomos/metabolismo , Inflamassomos/fisiologia , Interleucina-1beta/sangue , Lipopolissacarídeos , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/imunologia , Falência Hepática Aguda/metabolismo , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fator de Necrose Tumoral alfa/sangue
18.
Nat Protoc ; 15(10): 3284-3333, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32895525

RESUMO

Inflammasomes are multimeric heterogeneous mega-Dalton protein complexes that play key roles in the host innate immune response to infection and sterile insults. Assembly of the inflammasome complex following infection or injury begins with the oligomerization of the upstream inflammasome-forming sensor and proceeds through a multistep process of well-coordinated events and downstream effector functions. Together, these steps enable elegant experimental readouts with which to reliably assess the successful activation of the inflammasome complex and cell death. Here, we describe a comprehensive protocol that details several in vitro (in bone marrow-derived macrophages) and in vivo (in mice) strategies for activating the inflammasome and explain how to subsequently assess multiple downstream effects in parallel to unequivocally establish the activation status of the inflammasome and cell death pathways. Our workflow assesses inflammasome activation via the formation of the apoptosis-associated speck-like protein containing a CARD (ASC) speck; cleavage of caspase-1 and gasdermin D; release of IL-1ß, IL-18, caspase-1, and lactate dehydrogenase from the cell; and real-time analysis of cell death by imaging. Analyses take up to ~24 h to complete. Overall, our multifaceted approach provides a comprehensive and consistent protocol for assessing inflammasome activation and cell death.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Inflamassomos/metabolismo , Inflamassomos/fisiologia , Animais , Apoptose , Caspase 1/metabolismo , Morte Celular/fisiologia , Feminino , Imunidade Inata , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , L-Lactato Desidrogenase/metabolismo , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/metabolismo
19.
Life Sci ; 260: 118258, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32818542

RESUMO

Inflammation is a sophisticated biological tissue response to both extrinsic and intrinsic stimuli. Although the pathological aspects of inflammation are well appreciated, there are still rooms for understanding the physiological functions of the inflammation. Recent studies have focused on mechanisms, context and the role of physiological inflammation. Besides, there have been progress in the comprehension of commensal microbiota, immunometabolism, cancer and intracellular signaling events' roles that impact on the regulation of inflammation. Despite the fact that inflammatory responses are vital through tissue damage, understanding the mechanisms to turn off the finished or unnecessary inflammation is crucial for restoring homeostasis. Inflammation seems to be a smart process that acts like two edges of a sword, meaning that it has both protective and deleterious consequences. Knowing both edges and the regulation processes will help the future understanding and therapy for various diseases.


Assuntos
Inflamação/fisiopatologia , Animais , Homeostase , Humanos , Imunidade Inata/fisiologia , Inflamassomos/fisiologia , Microbiota/fisiologia , Fenômenos Fisiológicos da Nutrição , Transdução de Sinais , Baço/fisiopatologia
20.
PLoS One ; 15(8): e0237030, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32810141

RESUMO

Curative therapeutic options for a number of immunological disorders remain to be established, and approaches for identifying drug candidates are relatively limited. Furthermore, phenotypic screening methods using induced pluripotent stem cell (iPSC)-derived immune cells or hematopoietic cells need improvement. In the present study, using immortalized monocytic cell lines derived from iPSCs, we developed a high-throughput screening (HTS) system to detect compounds that inhibit IL-1ß secretion and NLRP3 inflammasome activation from activated macrophages. The iPSCs were generated from a patient with neonatal onset multisystem inflammatory disease (NOMID) as a model of a constitutively activated NLRP3 inflammasome. HTS of 4,825 compounds including FDA-approved drugs and compounds with known bioactivity identified 7 compounds as predominantly IL-1ß inhibitors. Since these compounds are known inflammasome inhibitors or derivatives of, these results prove the validity of our HTS system, which can be a versatile platform for identifying drug candidates for immunological disorders associated with monocytic lineage cells.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Linhagem Celular , Células Cultivadas , Síndromes Periódicas Associadas à Criopirina/imunologia , Síndromes Periódicas Associadas à Criopirina/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Inflamassomos/fisiologia , Inflamação/metabolismo , Interleucina-1beta , Macrófagos/metabolismo , Monócitos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA