Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.148
Filtrar
1.
Mol Biomed ; 5(1): 14, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38644450

RESUMO

NLRP inflammasomes are a group of cytosolic multiprotein oligomer pattern recognition receptors (PRRs) involved in the recognition of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) produced by infected cells. They regulate innate immunity by triggering a protective inflammatory response. However, despite their protective role, aberrant NLPR inflammasome activation and gain-of-function mutations in NLRP sensor proteins are involved in occurrence and enhancement of non-communicating autoimmune, auto-inflammatory, and neurodegenerative diseases. In the last few years, significant advances have been achieved in the understanding of the NLRP inflammasome physiological functions and their molecular mechanisms of activation, as well as therapeutics that target NLRP inflammasome activity in inflammatory diseases. Here, we provide the latest research progress on NLRP inflammasomes, including NLRP1, CARD8, NLRP3, NLRP6, NLRP7, NLRP2, NLRP9, NLRP10, and NLRP12 regarding their structural and assembling features, signaling transduction and molecular activation mechanisms. Importantly, we highlight the mechanisms associated with NLRP inflammasome dysregulation involved in numerous human auto-inflammatory, autoimmune, and neurodegenerative diseases. Overall, we summarize the latest discoveries in NLRP biology, their forming inflammasomes, and their role in health and diseases, and provide therapeutic strategies and perspectives for future studies about NLRP inflammasomes.


Assuntos
Inflamassomos , Proteínas NLR , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Proteínas NLR/metabolismo , Animais , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/metabolismo , Transdução de Sinais/imunologia , Imunidade Inata , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética
2.
Front Immunol ; 15: 1294898, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38660301

RESUMO

Human adenovirus type 7 (HAdV-7) is a significant viral pathogen that causes respiratory infections in children. Currently, there are no specific antiviral drugs or vaccines for children targeting HAdV-7, and the mechanisms of its pathogenesis remain unclear. The NLRP3 inflammasome-driven inflammatory cascade plays a crucial role in the host's antiviral immunity. Our previous study demonstrated that HAdV-7 infection activates the NLRP3 inflammasome. Building upon this finding, our current study has identified the L4 100 kDa protein encoded by HAdV-7 as the primary viral component responsible for NLRP3 inflammasome activation. By utilizing techniques such as co-immunoprecipitation, we have confirmed that the 100 kDa protein interacts with the NLRP3 protein and facilitates the assembly of the NLRP3 inflammasome by binding specifically to the NACHT and LRR domains of NLRP3. These insights offer a deeper understanding of HAdV-7 pathogenesis and contribute to the development of novel antiviral therapies.


Assuntos
Infecções por Adenovirus Humanos , Adenovírus Humanos , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Humanos , Adenovírus Humanos/imunologia , Adenovírus Humanos/fisiologia , Inflamassomos/metabolismo , Inflamassomos/imunologia , Infecções por Adenovirus Humanos/imunologia , Infecções por Adenovirus Humanos/metabolismo , Infecções por Adenovirus Humanos/virologia , Ligação Proteica , Células HEK293 , Proteínas Virais/metabolismo , Proteínas Virais/imunologia
3.
J Immunol ; 212(2): 335-345, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38047899

RESUMO

Although electric field-induced cell membrane permeabilization (electroporation) is used in a wide range of clinical applications from cancer therapy to cardiac ablation, the cellular- and molecular-level details of the processes that determine the success or failure of these treatments are poorly understood. Nanosecond pulsed electric field (nsPEF)-based tumor therapies are known to have an immune component, but whether and how immune cells sense the electroporative damage and respond to it have not been demonstrated. Damage- and pathogen-associated stresses drive inflammation via activation of cytosolic multiprotein platforms known as inflammasomes. The assembly of inflammasome complexes triggers caspase-1-dependent secretion of IL-1ß and in many settings a form of cell death called pyroptosis. In this study we tested the hypothesis that the nsPEF damage is sensed intracellularly by the NLRP3 inflammasome. We found that 200-ns PEFs induced aggregation of the inflammasome adaptor protein ASC, activation of caspase-1, and triggered IL-1ß release in multiple innate immune cell types (J774A.1 macrophages, bone marrow-derived macrophages, and dendritic cells) and in vivo in mouse skin. Efflux of potassium from the permeabilized cell plasma membrane was partially responsible for nsPEF-induced inflammasome activation. Based on results from experiments using both the NRLP3-specific inhibitor MCC950 and NLRP3 knockout cells, we propose that the damage created by nsPEFs generates a set of stimuli for the inflammasome and that more than one sensor can drive IL-1ß release in response to electrical pulse stimulation. This study shows, to our knowledge, for the first time, that PEFs activate the inflammasome, suggesting that this pathway alarms the immune system after treatment.


Assuntos
Inflamassomos , Interleucina-1beta , Macrófagos , Pele , Inflamassomos/imunologia , Interleucina-1beta/imunologia , Animais , Camundongos , Pele/imunologia , Células HEK293 , Humanos , Linhagem Celular , Gasderminas/imunologia , Estimulação Elétrica , Macrófagos/imunologia , Imunidade Inata/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
4.
Cell Rep ; 42(12): 113581, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38103201

RESUMO

Inflammasomes are multiprotein signaling complexes that activate the innate immune system. Canonical inflammasomes recruit and activate caspase-1, which then cleaves and activates IL-1ß and IL-18, as well as gasdermin D (GSDMD) to induce pyroptosis. In contrast, non-canonical inflammasomes, caspases-4/-5 (CASP4/5) in humans and caspase-11 (CASP11) in mice, are known to cleave GSDMD, but their role in direct processing of other substrates besides GSDMD has remained unknown. Here, we show that CASP4/5 but not CASP11 can directly cleave and activate IL-18. However, CASP4/5/11 can all cleave IL-1ß to generate a 27-kDa fragment that deactivates IL-1ß signaling. Mechanistically, we demonstrate that the sequence identity of the tetrapeptide sequence adjacent to the caspase cleavage site regulates IL-18 and IL-1ß recruitment and activation. Altogether, we have identified new substrates of the non-canonical inflammasomes and reveal key mechanistic details regulating inflammation that may aid in developing new therapeutics for immune-related disorders.


Assuntos
Caspases , Interleucina-18 , Interleucina-1beta , Caspases/genética , Caspases/imunologia , Interleucina-18/química , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-1beta/química , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Células RAW 264.7 , Células HEK293 , Células HeLa , Células THP-1 , Humanos , Inflamassomos/imunologia , Transdução de Sinais/genética , Proteólise , Ligação Proteica , Multimerização Proteica , Infecções por Salmonella/enzimologia , Infecções por Salmonella/imunologia
5.
Nature ; 623(7989): 1044-1052, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37993709

RESUMO

All nucleated cells express major histocompatibility complex I and interferon-γ (IFNγ) receptor1, but an epithelial cell-specific function of IFNγ signalling or antigen presentation by means of major histocompatibility complex I has not been explored. We show here that on sensing IFNγ, colonic epithelial cells productively present pathogen and self-derived antigens to cognate intra-epithelial T cells, which are critically located at the epithelial barrier. Antigen presentation by the epithelial cells confers extracellular ATPase expression in cognate intra-epithelial T cells, which limits the accumulation of extracellular adenosine triphosphate and consequent activation of the NLRP3 inflammasome in tissue macrophages. By contrast, antigen presentation by the tissue macrophages alongside inflammasome-associated interleukin-1α and interleukin-1ß production promotes a pathogenic transformation of CD4+ T cells into granulocyte-macrophage colony-stimulating-factor (GM-CSF)-producing T cells in vivo, which promotes colitis and colorectal cancer. Taken together, our study unravels critical checkpoints requiring IFNγ sensing and antigen presentation by epithelial cells that control the development of pathogenic CD4+ T cell responses in vivo.


Assuntos
Apresentação de Antígeno , Colo , Células Epiteliais , Interferon gama , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Colo/citologia , Colo/imunologia , Colo/patologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-1alfa/imunologia , Interleucina-1beta/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
6.
Front Biosci (Landmark Ed) ; 28(9): 210, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37796716

RESUMO

Inflammasomes are cytosolic multi-protein complexes that play an important role in the innate immune system, inducing cytokine maturation and pyroptosis. Trained immunity is the induction of memory in innate immune cells by epigenetic reprogramming due to repeated inflammatory stimuli that alter the inflammatory response and increase resistance to infection or disease. Although it is speculated that nucleotide-binding oligomerization domain (NOD), leucine-rich repeat (LRR), and the NLR family pyrin domain containing 3 (NLRP3) inflammasomes respond to various inflammatory stimuli and are associated with trained immunity, the exact relationship is still unclear. This paper aims to introduce data from recent research on the role of inflammasomes in trained immunity through cellular immunometabolic and epigenetic reprogramming. It also suggests a new therapeutic strategy for inflammatory diseases through the complementary regulation of inflammasomes and trained immunity.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Imunidade Treinada , Citocinas/imunologia , Inflamassomos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Piroptose/imunologia , Imunidade Treinada/imunologia , Humanos , Animais
7.
Front Cell Infect Microbiol ; 13: 1173505, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37465759

RESUMO

The inflammasome is a multiprotein complex that further regulates cell pyroptosis and inflammation by activating caspase-1. The assembly and activation of inflammasome are associated with a variety of diseases. Accumulative studies have shown that inflammasome is a key modulator of the host's defense response to viral infection. Indeed, it has been established that activation of inflammasome occurs during viral infection. At the same time, the host has evolved a variety of corresponding mechanisms to inhibit unnecessary inflammasome activation. Therefore, here, we review and summarize the latest research progress on the interaction between inflammosomes and viruses, highlight the assembly and activation of inflammosome in related cells after viral infection, as well as the corresponding molecular regulatory mechanisms, and elucidate the effects of this activation on virus immune escape and host innate and adaptive immune defenses. Finally, we also discuss the potential therapeutic strategies to prevent and/or ameliorate viral infection-related diseases via targeting inflammasomes and its products.


Assuntos
Interações entre Hospedeiro e Microrganismos , Inflamassomos , Viroses , Vírus , Humanos , Inflamassomos/imunologia , Viroses/imunologia , Viroses/terapia , Vírus/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Animais
8.
Antiviral Res ; 216: 105674, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37459896

RESUMO

Seasonal coronaviruses widely circulate in the global population, and severe complications can occur in specific vulnerable populations. Little is known on their pathogenic mechanisms and no approved treatment is available. Here, we present anecdotal evidence that the level of IL-1ß, a hallmark of inflammasome activation, appears elevated in a subset of seasonal coronavirus infected patients. We found that cultured human macrophages support the full life cycle of three cultivatable seasonal coronaviruses. Their infections effectively activate NLRP3 inflammasome activation through TLR4 ligation and NF-κB activation. This activation can be attenuated by specific pharmacological inhibitors and clinically used medications including dexamethasone and flufenamic acid. Interestingly, combination of antiviral and anti-inflammatory drugs simultaneously inhibit seasonal coronavirus-triggered inflammatory response and viral replication. Collectively, these findings show that the TLR4/NF-κB/NLRP3 axis drives seasonal coronavirus triggered-inflammatory response, which in turn represents a viable therapeutic target.


Assuntos
Infecções por Coronavirus , Inflamassomos , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Infecções por Coronavirus/imunologia , Humanos , Inflamassomos/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Estações do Ano , NF-kappa B/metabolismo , Linhagem Celular
10.
Cell ; 186(10): 2144-2159.e22, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37172565

RESUMO

Bats are special in their ability to live long and host many emerging viruses. Our previous studies showed that bats have altered inflammasomes, which are central players in aging and infection. However, the role of inflammasome signaling in combating inflammatory diseases remains poorly understood. Here, we report bat ASC2 as a potent negative regulator of inflammasomes. Bat ASC2 is highly expressed at both the mRNA and protein levels and is highly potent in inhibiting human and mouse inflammasomes. Transgenic expression of bat ASC2 in mice reduced the severity of peritonitis induced by gout crystals and ASC particles. Bat ASC2 also dampened inflammation induced by multiple viruses and reduced mortality of influenza A virus infection. Importantly, it also suppressed SARS-CoV-2-immune-complex-induced inflammasome activation. Four key residues were identified for the gain of function of bat ASC2. Our results demonstrate that bat ASC2 is an important negative regulator of inflammasomes with therapeutic potential in inflammatory diseases.


Assuntos
Proteínas Reguladoras de Apoptose , Quirópteros , Inflamassomos , Ribonucleoproteínas , Viroses , Animais , Humanos , Camundongos , Proteínas Reguladoras de Apoptose/metabolismo , Quirópteros/imunologia , COVID-19 , Inflamassomos/imunologia , Ribonucleoproteínas/metabolismo , SARS-CoV-2 , Viroses/imunologia , Fenômenos Fisiológicos Virais
11.
J Virol ; 97(6): e0058923, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37255428

RESUMO

The inflammasome pathway is a critical early response mechanism of the host that detects pathogens, initiates the production of inflammatory cytokines, and recruits effector cells to the infection site. Nonetheless, the mechanism of inflammasome activation in coronavirus infection and its biological functions in host defense remain unclear. Transmissible gastroenteritis virus (TGEV), a member of the genus Alphacoronavirus, is a significant pathogen that mainly infects piglets and causes intestinal inflammation and inflammatory cell infiltration. Here, we investigated the mechanism of inflammasome activation in intestinal epithelial cells (IECs) infected with TGEV. We observed a substantial increase in interleukin 1ß (IL-1ß) and IL-18 levels in both IECs and TGEV-infected porcine intestinal tissues. Furthermore, TGEV infection resulted in increased activation of caspase-1 and the NLRP1 (NOD-like receptor [NLR]-containing pyrin domain [PYD]) inflammasome. Our findings revealed that TGEV infection impeded the interaction between porcine NLRP1 (pNLRP1) and porcine dipeptidyl peptidases 9 (pDPP9), yet it did not reduce the expression of pDPP9. Importantly, the ZU5 domain, not the function-to-find domain (FIIND) reported in human NLRP1, was identified as the minimal domain of pNLRP1 for pDPP9 binding. In addition, the robust type I IFN expression induced by TGEV infection also upregulated pNLRP1 expression and pNLRP1 itself acts as an interferon-stimulated gene to counteract TGEV infection. Our data demonstrate that pNLRP1 has antiviral capabilities against coronavirus infection, which highlights its potential as a novel therapeutic target for coronavirus antiviral therapy. IMPORTANCE Coronavirus primarily targets the epithelial cells of the respiratory and gastrointestinal tracts, leading to damage in both humans and animals. NLRP1 is a direct sensor for RNA virus infection which is highly expressed in epithelial barrier tissues. However, until recently, the precise molecular mechanisms underlying its activation in coronavirus infection and subsequent downstream events remained unclear. In this study, we demonstrate that the alphacoronavirus TGEV induces the production of IL-1ß and IL-18 and upregulates the expression of pNLRP1. Furthermore, we found that pNLRP1 can serve as an interferon-stimulated gene (ISG) to inhibit the infection of enterovirus TGEV. Our research highlights the crucial role of NLRP1 as a regulator of innate immunity in TGEV infection and shows that it may serve as a potential therapeutic target for the treatment of coronavirus infection.


Assuntos
Gastroenterite Suína Transmissível , Inflamassomos , Proteínas NLR , Vírus da Gastroenterite Transmissível , Animais , Inflamassomos/imunologia , Interferon Tipo I , Interleucina-18 , Proteínas NLR/imunologia , Suínos , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/transmissão
12.
Biol Res Nurs ; 25(2): 267-281, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36207143

RESUMO

BACKGROUND: Gout is triggered by high urate levels and causes inflammation, pain, and an impaired quality of life. Immersion in water at 20-30°C reduces inflammation and pain in arthritis. Yet, relationships of immersion in water at 20-30°C with urate levels and the nucleotide-binding domain (NOD)-like receptor protein 1 (NLRP1) inflammasome have never been clarified. OBJECTIVES: We aimed to investigate the effects of immersion in water at 20-30°C on urate levels, the NLRP1 inflammasome, pain, and quality of life among acute gout patients. METHODS: A community-based randomized control trial design was used with 2 parallel-intervention groups: immersion in water at 20-30°C (20 min/day for 4 weeks) group and a control group. In total, 76 eligible participants in Tomohon City, Indonesia, were assigned using block randomization. We analyze the results (coef. ß) and 95% confidence intervals (CIs) using a generalized estimating equation model. We analyzed mediating effects using a path analysis. RESULTS: Significant pain alleviation (ß = -2.06 [95% CI = -2.67∼-1.45]; ß = -2.42 [95% CI = -2.97∼-1.87]) and improved quality of life (ß = 5.34 [95% CI = 3.12-7.57]; ß = 9.93 [95% CI = 7.02-12.83]) were detected at 2 and 4 weeks of follow-up compared to the pre-test and control group. Urate levels (ß = -0.34 [95% CI = -0.52∼-0.16]) were reduced at the 2-week follow-up, but there was no significant change in the NLRP1 inflammasome compared to the pre-test and control group after immersion in water at 20-30°C. Both the NLRP1 inflammasome (ß = -0.48 [95% CI = -0.63∼-0.34]); water 0.01) and urate levels (ß = -0.11 [95% CI = -0.24∼-0.03]; p < 0.01) had partial indirect (mediating) effects on the link between immersion in water at 20-30°C and pain at the 4-week follow-up. CONCLUSIONS: Immersion in water at 20-30°C significantly decreased pain and increased the quality of life. Immersion in water at 20-30°C mediated NLRP1 and urate levels to decrease pain, although it had no significant effect on the NLRP1 inflammasome concentration after 4 weeks of follow-up and reduced urate levels only at 2 weeks after immersion in water at 20-30°C.


Assuntos
Gota , Inflamassomos , Inflamação , Manejo da Dor , Dor , Humanos , Gota/complicações , Gota/genética , Gota/imunologia , Gota/terapia , Imersão , Indonésia , Inflamassomos/genética , Inflamassomos/imunologia , Inflamação/genética , Inflamação/imunologia , Dor/genética , Dor/imunologia , Manejo da Dor/métodos , Qualidade de Vida , Temperatura , Ácido Úrico/efeitos adversos , Ácido Úrico/análise , Água , Biomarcadores
13.
Mol Ther ; 31(3): 890-908, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36566348

RESUMO

Mesenchymal stem cells (MSCs) are ubiquitous multipotent cells that exhibit significant therapeutic potentials in a variety of disorders. Nevertheless, their clinical efficacy is limited owing to poor survival, low rate of engraftment, and impaired potency upon transplantation. Spheroidal three-dimensional (3D) culture of MSCs (MSC3D) has been proven to better preserve their in vivo functional properties. However, the molecular mechanisms underlying the improvement in MSC function by spheroid formation are not clearly understood. NLRP3 inflammasomes, a key component of the innate immune system, have recently been shown to play a role in cell fate decision of MSCs. The present study examined the role of NLRP3 inflammasomes in the survival and potency of MSC spheroids. We found that MSC3D led to decreased activation of NLRP3 inflammasomes through alleviation of ER stress in an autophagy-dependent manner. Importantly, downregulation of NLRP3 inflammasomes signaling critically contributes to the enhanced survival rate in MSC3D through modulation of pyroptosis and apoptosis. The critical role of NLRP3 inflammasome suppression in the enhanced therapeutic efficacy of MSC spheroids was further confirmed in an in vivo mouse model of DSS-induced colitis. These findings suggest that 3D culture confers survival and functional advantages to MSCs by suppressing NLRP3 inflammasome activation.


Assuntos
Colite , Inflamassomos , Células-Tronco Mesenquimais , Animais , Camundongos , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Inflamassomos/genética , Inflamassomos/imunologia , Células-Tronco Mesenquimais/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Transdução de Sinais , Técnicas de Cultura de Células em Três Dimensões
14.
FEBS J ; 290(6): 1549-1562, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36181338

RESUMO

Sepsis often causes cell death via pyroptosis and hence results in septic cardiomyopathy. Triggering receptors expressed in myeloid cells-1 (TREM-1) may initiate cellular cascade pathways and, in turn, induce cell death and vital organ dysfunction in sepsis, but the evidence is limited. We set to investigate the role of TREM-1 on nucleotide-binding oligomerization domain-like receptors with pyrin domain-3 (NLRP3) inflammasome activation and cardiomyocyte pyroptosis in sepsis models using cardiac cell line (HL-1) and mice. In this study, TREM-1 was found to be significantly increased in HL-1 cells challenged with lipopolysaccharide (LPS). Pyroptosis was also significantly increased in the HL-1 cells challenged with lipopolysaccharide and an NLRP3 inflammasome activator, nigericin. The close interaction between TREM-1 and structural maintenance of chromosome 4 (SMC4) was also identified. Furthermore, inhibition of TREM-1 or SMC4 prevented the upregulation of NLRP3 and decreased Gasdermin-D, IL-1ß and caspase-1 cleavage. In mice subjected to caecal ligation and puncture, the TREM-1 inhibitor LR12 decreased the expression of NLRP3 and attenuated cardiomyocyte pyroptosis, leading to improved cardiac function and prolonged survival of septic mice. Our work demonstrates that, under septic conditions, TREM-1 plays a critical role in cardiomyocyte pyroptosis. Targeting TREM-1 and its associated molecules may therefore lead to novel therapeutic treatments for septic cardiomyopathy.


Assuntos
Inflamassomos , Miócitos Cardíacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Sepse , Receptor Gatilho 1 Expresso em Células Mieloides , Animais , Humanos , Camundongos , Adenosina Trifosfatases/imunologia , Cardiomiopatias/etiologia , Cardiomiopatias/genética , Cardiomiopatias/imunologia , Caspase 1/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/imunologia , Cromossomos Humanos Par 4/imunologia , Inflamassomos/agonistas , Inflamassomos/genética , Inflamassomos/imunologia , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/farmacologia , Células Mieloides/imunologia , Miócitos Cardíacos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/agonistas , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Piroptose/genética , Piroptose/imunologia , Sepse/complicações , Sepse/genética , Sepse/imunologia , Receptor Gatilho 1 Expresso em Células Mieloides/antagonistas & inibidores , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Receptor Gatilho 1 Expresso em Células Mieloides/imunologia
15.
Int Immunopharmacol ; 113(Pt A): 109375, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36461592

RESUMO

BACKGROUND: Recent studies have uncovered that hyperuricemia (HUA) leads to cognitive deficits, which are accompanied by neuronal damage and neuroinflammation. Here, we aim to explore the role of methyltransferase-like 3 (METTL3) in HUA-mediated neuronal apoptosis and microglial inflammation. METHODS: A HUA mouse model was constructed. The spatial memory ability of the mice was assessed by the Morris water maze experiment (MWM), and neuronal apoptosis was analyzed by the TdT-mediated dUTP nick end labeling (TUNEL) assay. Besides, enzyme-linked immunosorbent assay (ELISA) was utilized to measure the contents of inflammatory factors (IL-1ß, IL-6, and TNF-α) and oxidative stress markers (MDA, SOD, and CAT) in the serum of mice. In vitro, the mouse hippocampal neuron (HT22) and microglia (BV2) were treated with uric acid (UA). Flow cytometry was applied to analyze HT22 and BV2 cell apoptosis, and ELISA was conducted to observe neuroinflammation and oxidative stress. In addition, the expression of MyD88, p-NF-κB, NF-κB, NLRP3, ASC and Caspase1 was determined by Western blot. RESULTS: METTL3 and miR-124-3p were down-regulated, while the MyD88-NF-κB pathway was activated in the HUA mouse model. UA treatment induced neuronal apoptosis in HT22 and stimulated microglial activation in BV2. Overexpressing METTL3 alleviated HT22 neuronal apoptosis and resisted the release of inflammatory cytokines and oxidative stress mediators in BV2 cells. METTL3 repressed MyD88-NF-κB and NLRP3-ASC-Caspase1 inflammasome. In addition, METTL3 overexpression enhanced miR-124-3p expression, while METTL3 knockdown aggravated HT22 cell apoptosis and BV2 cell overactivation. CONCLUSION: METTL3 improves neuronal apoptosis and microglial activation in the HUA model by choking the MyD88/NF-κB pathway and up-regulating miR-124-3p.


Assuntos
Disfunção Cognitiva , Hiperuricemia , Inflamassomos , Metiltransferases , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/imunologia , Caspase 1/genética , Caspase 1/imunologia , Células Cultivadas , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/imunologia , Modelos Animais de Doenças , Hiperuricemia/complicações , Hiperuricemia/genética , Hiperuricemia/imunologia , Inflamassomos/genética , Inflamassomos/imunologia , Metiltransferases/genética , Metiltransferases/imunologia , MicroRNAs/genética , MicroRNAs/imunologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/imunologia , Sistema Nervoso/fisiopatologia , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/genética , Doenças Neuroinflamatórias/imunologia , NF-kappa B , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Ácido Úrico/administração & dosagem , Ácido Úrico/efeitos adversos , Ácido Úrico/farmacologia
16.
Respir Res ; 23(1): 326, 2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36463179

RESUMO

BACKGROUND: Bacterial pneumonia is a major risk factor for acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Pseudomonas aeruginosa (PA), an opportunistic pathogen with an increasing resistance acquired against multiple drugs, is one of the main causative agents of ALI and ARDS in diverse clinical settings. Given the anti-inflammatory role of the cannabinoid-2 receptor (CB2R), the effect of CB2R activation in the regulation of PA-induced ALI and inflammation was tested in a mouse model as an alternative to conventional antibiotic therapy. METHODS: In order to activate CB2R, a selective synthetic agonist, JWH133, was administered intraperitoneally (i.p.) to C57BL/6J mice. Furthermore, SR144528 (a selective CB2R antagonist) was administered in combination with JWH133 to test the specificity of the CB2R-mediated effect. PA was administered intratracheally (i.t.) for induction of pneumonia in mice. At 24 h after PA exposure, lung mechanics were measured using the FlexiVent system. The total cell number, protein content, and neutrophil population in the bronchoalveolar lavage fluid (BALF) were determined. The bacterial load in the whole lung was also measured. Lung injury was evaluated by histological examination and PA-induced inflammation was assessed by measuring the levels of BALF cytokines and chemokines. Neutrophil activation (examined by immunofluorescence and immunoblot) and PA-induced inflammatory signaling (analyzed by immunoblot) were also studied. RESULTS: CB2R activation by JWH133 was found to significantly reduce PA-induced ALI and the bacterial burden. CB2R activation also suppressed the PA-induced increase in immune cell infiltration, neutrophil population, and inflammatory cytokines. These effects were abrogated by a CB2R antagonist, SR144528, further confirming the specificity of the CB2R-mediated effects. CB2R-knock out (CB2RKO) mice had a significantly higher level of PA-induced inflammation as compared to that in WT mice. CB2R activation diminished the excess activation of neutrophils, whereas mice lacking CB2R had elevated neutrophil activation. Pharmacological activation of CB2R significantly reduced the PA-induced NF-κB and NLRP3 inflammasome activation, whereas CB2KO mice had elevated NLRP3 inflammasome. CONCLUSION: Our findings indicate that CB2R activation ameliorates PA-induced lung injury and inflammation, thus paving the path for new therapeutic avenues against PA pneumonia.


Assuntos
Lesão Pulmonar Aguda , Canabinoides , Inflamação , Infecções por Pseudomonas , Receptor CB2 de Canabinoide , Animais , Camundongos , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/prevenção & controle , Canabinoides/farmacologia , Canabinoides/uso terapêutico , Citocinas , Inflamassomos/genética , Inflamassomos/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/prevenção & controle , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Pseudomonas aeruginosa , Receptores de Canabinoides , Síndrome do Desconforto Respiratório , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/imunologia , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/prevenção & controle , Modelos Animais de Doenças
17.
J Ovarian Res ; 15(1): 126, 2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36463191

RESUMO

The survival of ovary granulosa cells (GC) is critical in the initiation and progression of polycystic ovary syndrome (PCOS) in females. Here, we found that the PCOS process is accompanied by massive GC pyroptosis resulting from Caspase-1 inflammasome activation. Administration of plumbagin, an effective compound isolated from plant medicine, can prevent the pyroptosis of GC and the onset of PCOS. Mechanistic study indicates the over-activation of the inflammasome in GC is due to the upregulation of WTAP, a key regulator of the RNA N6-methylase complex. WTAP mediates the mRNA N6-methylation of NLRP3 inflammasome component ASC and enhances ASC RNA stability, which results in the overactivation of the inflammasome in GCs from the PCOS model. Plumbagin treatment suppresses the WTAP-mediated N6-methylation of ASC mRNA and reduces the pyroptosis of GCs. This study supports the profound potential of plumbagin in PCOS treatment.


Assuntos
Células da Granulosa , Naftoquinonas , Síndrome do Ovário Policístico , Piroptose , Feminino , Humanos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/imunologia , Inflamassomos/genética , Inflamassomos/imunologia , Metilação/efeitos dos fármacos , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/imunologia , Piroptose/efeitos dos fármacos , Piroptose/genética , Piroptose/imunologia , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/imunologia , RNA Mensageiro , Naftoquinonas/imunologia , Naftoquinonas/farmacologia , Naftoquinonas/uso terapêutico
18.
Front Immunol ; 13: 832306, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091026

RESUMO

Neutrophils play major roles against bacteria and fungi infections not only due to their microbicide properties but also because they release mediators like Interleukin-1 beta (IL-1ß) that contribute to orchestrate the inflammatory response. This cytokine is a leaderless protein synthesized in the cytoplasm as a precursor (pro-IL-1ß) that is proteolytically processed to its active isoform and released from human neutrophils by secretory autophagy. In most myeloid cells, pro-IL-1ß is processed by caspase-1 upon inflammasome activation. Here we employed neutrophils from both healthy donors and patients with a gain-of-function (GOF) NLRP3-mutation to dissect IL-1ß processing in these cells. We found that although caspase-1 is required for IL-1ß secretion, it undergoes rapid inactivation, and instead, neutrophil serine proteases play a key role in pro-IL-1ß processing. Our findings bring to light distinctive features of the regulation of caspase-1 activity in human neutrophils and reveal new molecular mechanisms that control human neutrophil IL-1ß secretion.


Assuntos
Autofagia , Caspase 1 , Interleucina-1beta , Neutrófilos , Serina Proteases , Autofagia/genética , Autofagia/imunologia , Caspase 1/genética , Caspase 1/metabolismo , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Neutrófilos/enzimologia , Neutrófilos/imunologia , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Serina Proteases/genética , Serina Proteases/imunologia
19.
Dis Markers ; 2022: 7172583, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35968506

RESUMO

Objective: MicroRNAs have been revealed to be involved in the development of atherosclerosis. The present study is aimed at exploring the potential of miR-99a-5p as a therapy for atherosclerosis. We suspected that miR-99a-5p might inhibit NLRP3 inflammasome activation and promote macrophage autophagy via constraining mTOR, therefore, alleviating atherosclerosis. Methods: The cell viability in ox-LDL-induced THP-1 macrophages was assessed by CCK-8 assay. Bioinformatic analysis was used to predict the target genes of miR-99a-5p. The binding between miR-99a-5p and mTOR was confirmed by luciferase reporter assay. In vivo, a high-fat-diet-induced atherosclerosis model was established in apolipoprotein E knockout mice. Hematoxylin-eosin, oil red O, and Sirius red staining were performed for the determination of atherosclerotic lesions. MTOR and associated protein levels were detected by Western blot analysis. Results: miR-99a-5p inhibited NLRP3 inflammasome activation and promoted macrophage autophagy by targeting mTOR. Enforced miR-99a-5p significantly reduced the levels of inflammasome complex and inflammatory cytokines. Furthermore, miR-99a-5p overexpression inhibited the expression of mTOR, whereas mTOR overexpression reversed the trend of the above behaviors. In vivo, the specific overexpression of miR-99a-5p significantly reduced atherosclerotic lesions, accompanied by a significant downregulation of autophagy marker CD68 protein expression. Conclusion: We demonstrated for the first time that miR-99a-5p may be considered a therapy for atherosclerosis. The present study has revealed that miR-99a-5p might inhibit NLRP3 inflammasome activation and promote macrophage autophagy by targeting mTOR, therefore, alleviating atherosclerosis.


Assuntos
Aterosclerose , MicroRNAs , Proteína 3 que Contém Domínio de Pirina da Família NLR , Serina-Treonina Quinases TOR , Animais , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/terapia , Autofagia , Inflamassomos/genética , Inflamassomos/imunologia , Lipoproteínas LDL , Macrófagos/imunologia , Camundongos , MicroRNAs/genética , MicroRNAs/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia
20.
Cell Mol Gastroenterol Hepatol ; 14(4): 751-767, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35787975

RESUMO

BACKGROUND & AIMS: Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease worldwide. The NLRP3 inflammasome, a platform for caspase-1 activation and release of interleukin 1ß, is increasingly recognized in the induction of inflammation and liver fibrosis during NAFLD. However, the cell-specific contribution of NLRP3 inflammasome activation in NAFLD remains unknown. METHODS: To investigate the role of NLRP3 inflammasome activation in hepatocytes, hepatic stellate cells (HSCs) and myeloid cells, a conditional Nlrp3 knock-out mouse was generated and bred to cell-specific Cre mice. Both acute and chronic liver injury models were used: lipopolysaccharide/adenosine-triphosphate to induce in vivo NLRP3 activation, choline-deficient, L-amino acid-defined high-fat diet, and Western-type diet to induce fibrotic nonalcoholic steatohepatitis (NASH). In vitro co-culture studies were performed to dissect the crosstalk between myeloid cells and HSCs. RESULTS: Myeloid-specific deletion of Nlrp3 blunted the systemic and hepatic increase in interleukin 1ß induced by lipopolysaccharide/adenosine-triphosphate injection. In the choline-deficient, L-amino acid-defined high-fat diet model of fibrotic NASH, myeloid-specific Nlrp3 knock-out but not hepatocyte- or HSC-specific knock-out mice showed significant reduction in inflammation independent of steatosis development. Moreover, myeloid-specific Nlrp3 knock-out mice showed ameliorated liver fibrosis and decreased HSC activation. These results were validated in the Western-type diet model. In vitro co-cultured studies with human cell lines demonstrated that HSC can be activated by inflammasome stimulation in monocytes, and this effect was significantly reduced if NLRP3 was downregulated in monocytes. CONCLUSIONS: The study provides new insights in the cell-specific role of NLRP3 in liver inflammation and fibrosis. NLRP3 inflammasome activation in myeloid cells was identified as crucial for the progression of NAFLD to fibrotic NASH. These results may have implications for the development of cell-specific strategies for modulation of NLRP3 activation for treatment of fibrotic NASH.


Assuntos
Inflamassomos , Cirrose Hepática , Células Mieloides , Proteína 3 que Contém Domínio de Pirina da Família NLR , Hepatopatia Gordurosa não Alcoólica , Adenosina , Aminoácidos , Animais , Caspases , Colina , Hepatite/genética , Hepatite/imunologia , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Inflamação , Interleucina-1beta/imunologia , Lipopolissacarídeos , Cirrose Hepática/genética , Cirrose Hepática/imunologia , Camundongos , Camundongos Knockout , Células Mieloides/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/imunologia , Polifosfatos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...