Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.240
Filtrar
1.
Signal Transduct Target Ther ; 9(1): 245, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39300122

RESUMO

Pyroptosis is a type of programmed cell death characterized by cell swelling and osmotic lysis, resulting in cytomembrane rupture and release of immunostimulatory components, which play a role in several pathological processes. Significant cellular responses to various stimuli involve the formation of inflammasomes, maturation of inflammatory caspases, and caspase-mediated cleavage of gasdermin. The function of pyroptosis in disease is complex but not a simple angelic or demonic role. While inflammatory diseases such as sepsis are associated with uncontrollable pyroptosis, the potent immune response induced by pyroptosis can be exploited as a therapeutic target for anti-tumor therapy. Thus, a comprehensive review of the role of pyroptosis in disease is crucial for further research and clinical translation from bench to bedside. In this review, we summarize the recent advancements in understanding the role of pyroptosis in disease, covering the related development history, molecular mechanisms including canonical, non-canonical, caspase 3/8, and granzyme-mediated pathways, and its regulatory function in health and multiple diseases. Moreover, this review also provides updates on promising therapeutic strategies by applying novel small molecule inhibitors and traditional medicines to regulate pyroptosis. The present dilemmas and future directions in the landscape of pyroptosis are also discussed from a clinical perspective, providing clues for scientists to develop novel drugs targeting pyroptosis.


Assuntos
Piroptose , Piroptose/genética , Humanos , Inflamassomos/metabolismo , Inflamassomos/genética , Inflamassomos/imunologia , Granzimas/genética , Granzimas/metabolismo , Sepse/genética , Sepse/patologia , Sepse/metabolismo , Sepse/imunologia , Caspase 8/genética , Caspase 8/metabolismo , Neoplasias/genética , Neoplasias/patologia , Neoplasias/metabolismo , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Transdução de Sinais
2.
Dev Comp Immunol ; 161: 105263, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39265857

RESUMO

Chinese giant salamander (Andrias davidianus) is the largest extant urodela species and has unique evolutionary position. Studying the immune system of Chinese giant salamander contributes to understanding the evolution of immune systems of vertebrates. The NLR-related protein 3 (NLRP3) inflammasome comprised of NLRP3, ASC and caspase-1 play important roles in the host innate immunity. However, little is know about the NLRP3 inflammasome components in Chinese giant salamander. In this study, the NLRP3, apoptosis-associated speck-like protein (ASC) and caspase-1 (adaNLRP3, adaASC and adaCaspase-1) were characterized from Chinese giant salamander. The proteins of these three genes shared similar motifs and structures with their mammalian counterparts, with a PYD motif, a nucleotide-binding domain (NACHT) motif, and four leucine-rich repeat domain (LRR) motifs identified in adaNLRP3, a pyrin domain (PYD) motif and a caspase recruitment domain (CARD) motif in adaASC, and a CARD motif and a CASc motif in adaCaspase-1. These three genes were constitutively expressed in the skin, heart, lung, kidney, muscle, brain, spleen, and liver of Chinese giant salamander. Following Aeromonas hydrophia infection, all the three genes were up-regulated in various tissues. Molecular docking analysis revealed that the key residues involved in forming the adaNLRP3/adaASC complex were located in the PYD motifs, and that involved in forming the adaASC/adaCaspase-1 complex were located in the CARD motifs. Further analysis revealed that the hydrogen bonds and salt bridges had crucial roles in the formation of adaNLRP3/acaASC and adaASC/adaCaspase-1 complexes. To the best of our knowledge, this is the first report on the NLRP3 inflammasome components in Chinese giant salamander which will be helpful in further understanding the function of the NLRP3 inflammasome and in elucidating its role in the immune response to microbes.


Assuntos
Imunidade Inata , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Urodelos , Animais , Urodelos/imunologia , Urodelos/genética , Inflamassomos/metabolismo , Inflamassomos/imunologia , Inflamassomos/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Espécies em Perigo de Extinção , Proteínas de Anfíbios/metabolismo , Proteínas de Anfíbios/genética , Caspase 1/metabolismo , Caspase 1/genética , Filogenia
3.
Pharmacol Res ; 208: 107411, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39270948

RESUMO

Despite considerable improvements in understanding the biology of glioblastoma (GB), it still remains the most lethal type of brain tumor in adults. The role of innate immune cells in the development of GB was recently described. In particular, the tumor-immune cell interactions are thought to be critical in enabling tumor tolerance and even protection against therapeutics. Interestingly, the GB cells express proteins belonging to the family of intracellular pattern-recognition receptors, namely the NOD-like receptors (NLRs). Their activation may trigger the formation of the inflammasome complex leading to the secretion of mature IL-1ß and IL-18 and thus resulting in cell death. Intrudingly, the expression of most NLRs was found to be correlated with tumor progression and poor prognosis. We speculate that recognizing the role of NOD-like receptors in GB has the potential to improve the effectiveness of diagnostic tools and prognosis, while also encouraging the development of novel precision medicine-based therapies.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/metabolismo , Glioblastoma/imunologia , Glioblastoma/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/genética , Prognóstico , Animais , Proteínas NLR/metabolismo , Inflamassomos/metabolismo , Inflamassomos/imunologia
4.
Cells ; 13(17)2024 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-39272998

RESUMO

There is an urgent need for effective disease-modifying therapeutic interventions for Alzheimer's disease (AD)-the most prevalent cause of dementia with a profound socioeconomic burden. Most clinical trials targeting the classical hallmarks of this disease-ß-amyloid plaques and neurofibrillary tangles-failed, showed discrete clinical effects, or were accompanied by concerning side effects. There has been an ongoing search for novel therapeutic targets. Neuroinflammation, now widely recognized as a hallmark of all neurodegenerative diseases, has been proven to be a major contributor to AD pathology. Here, we summarize the role of neuroinflammation in the pathogenesis and progression of AD and discuss potential targets such as microglia, TREM2, the complement system, inflammasomes, and cytosolic DNA sensors. We also present an overview of ongoing studies targeting specific innate immune system components, highlighting the progress in this field of drug research while bringing attention to the delicate nature of innate immune modulations in AD.


Assuntos
Doença de Alzheimer , Imunidade Inata , Doença de Alzheimer/imunologia , Humanos , Animais , Microglia/imunologia , Microglia/metabolismo , Inflamassomos/metabolismo , Inflamassomos/imunologia , Terapia de Alvo Molecular , Glicoproteínas de Membrana , Receptores Imunológicos
5.
Cytokine Growth Factor Rev ; 79: 52-65, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39227243

RESUMO

The activation of immune cells by pro-inflammatory or immunosuppressive stimuli is followed by the secretion of immunoregulatory cytokines which serve as messengers to activate the immune response in target cells. Although the mechanisms that control the secretion of cytokines by immune cells are not yet fully understood, several key aspects of this process have recently emerged. This review focuses on cytokine release via exocytosis and highlights the routes of cytokine trafficking leading to constitutive and regulated secretion as well as the impact of sorting receptors on this process. We discuss the involvement of cytoskeletal rearrangements in vesicular transport, secretion, and formation of immunological synapses. Finally, we describe the non-classical pathways of cytokine release that are independent of vesicular ER-Golgi transport. Instead, these pathways are based on processing by inflammasome or autophagic mechanisms. Ultimately, understanding the molecular mechanisms behind cytokine release may help to identify potential therapeutic targets in diseases associated with altered immune responses.


Assuntos
Citocinas , Exocitose , Humanos , Citocinas/imunologia , Citocinas/metabolismo , Animais , Exocitose/imunologia , Inflamassomos/imunologia , Autofagia/imunologia , Sinapses Imunológicas/imunologia , Transporte Proteico , Complexo de Golgi/metabolismo
7.
J Immunol ; 213(4): 407-417, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39102612

RESUMO

The Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome has been the most distinctive polymer protein complex. After recognizing the endogenous and exogenous danger signals, NLRP3 can cause inflammation by pyroptosis and secretion of mature, bioactive forms of IL-1ß and IL-18. The NLRP3 inflammasome is essential in the genesis and progression of infectious illnesses. Herein, we provide a comprehensive review of the NLRP3 inflammasome in infectious diseases, focusing on its two-sided effects. As an essential part of host defense with a protective impact, abnormal NLRP3 inflammasome activation, however, result in a systemic high inflammatory response, leading to subsequent damage. In addition, scientific evidence of small molecules, biologics, and phytochemicals acting on the NLRP3 inflammasome has been reviewed. We believe that the NLRP3 inflammasome helps us understand the pathological mechanism of different stages of infectious diseases and that inhibitors targeting the NLRP3 inflammasome will become a new and valuable research direction for the treatment of infectious diseases.


Assuntos
Doenças Transmissíveis , 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 , Inflamassomos/imunologia , Inflamassomos/metabolismo , Animais , Doenças Transmissíveis/imunologia , Doenças Transmissíveis/metabolismo , Inflamação/imunologia , Interleucina-1beta/metabolismo , Interleucina-1beta/imunologia
8.
Cell Host Microbe ; 32(8): 1427-1443.e8, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39094584

RESUMO

Prokaryotes have evolved a multitude of defense systems to protect against phage predation. Some of these resemble eukaryotic genes involved in antiviral responses. Here, we set out to systematically project the current knowledge of eukaryotic-like antiviral defense systems onto prokaryotic genomes, using Pseudomonas aeruginosa as a model organism. Searching for phage defense systems related to innate antiviral genes from vertebrates and plants, we uncovered over 450 candidates. We validated six of these phage defense systems, including factors preventing viral attachment, R-loop-acting enzymes, the inflammasome, ubiquitin pathway, and pathogen recognition signaling. Collectively, these defense systems support the concept of deep evolutionary links and shared antiviral mechanisms between prokaryotes and eukaryotes.


Assuntos
Pseudomonas aeruginosa , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/virologia , Imunidade Inata , Bacteriófagos/genética , Bacteriófagos/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética , Animais , Evolução Molecular , Inflamassomos/imunologia , Inflamassomos/genética , Eucariotos/virologia , Eucariotos/genética , Eucariotos/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Evolução Biológica , Plantas/imunologia , Plantas/virologia , Plantas/microbiologia
9.
Int J Mol Sci ; 25(15)2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39125766

RESUMO

Tuberculosis (TB), caused by Mycobacterium tuberculosis (M. tb), is a major global health issue, with around 10 million new cases annually. Advances in TB immunology have improved our understanding of host signaling pathways, leading to innovative therapeutic strategies. Inflammasomes, protein complexes organized by cytosolic pattern recognition receptors (PRRs), play a crucial role in the immune response to M. tb by activating caspase 1, which matures proinflammatory cytokines IL1ß and IL18. While inflammation is necessary to fight infection, excessive or dysregulated inflammation can cause tissue damage, highlighting the need for precise inflammasome regulation. Drug-resistant TB strains have spurred research into adjunctive host-directed therapies (HDTs) that target inflammasome pathways to control inflammation. Canonical and non-canonical inflammasome pathways can trigger excessive inflammation, leading to immune system exhaustion and M. tb spread. Novel HDT interventions can leverage precision medicine by tailoring treatments to individual inflammasome responses. Studies show that medicinal plant derivatives like silybin, andrographolide, and micheliolide and small molecules such as OLT1177, INF39, CY-09, JJ002, Ac-YVAD-cmk, TAK-242, and MCC950 can modulate inflammasome activation. Molecular tools like gene silencing and knockouts may also be used for severe TB cases. This review explores these strategies as potential adjunctive HDTs in fighting TB.


Assuntos
Inflamassomos , Mycobacterium tuberculosis , Tuberculose , Humanos , Inflamassomos/metabolismo , Inflamassomos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , Animais , Antituberculosos/uso terapêutico , Antituberculosos/farmacologia , Transdução de Sinais , Interações Hospedeiro-Patógeno/imunologia
10.
Front Immunol ; 15: 1416275, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39139560

RESUMO

The intermediate filament vimentin is present in immune cells and is implicated in proinflammatory immune responses. Whether and how it supports antimicrobial activities of neutrophils are not well established. Here, we developed an immortalized neutrophil model to examine the requirement of vimentin. We demonstrate that vimentin restricts the production of proinflammatory cytokines and reactive oxygen species (ROS), but enhances phagocytosis and swarming. We observe that vimentin is dispensable for neutrophil extracellular trap (NET) formation, degranulation, and inflammasome activation. Moreover, gene expression analysis demonstrated that the presence of vimentin was associated with changes in expression of multiple genes required for mitochondrial function and ROS overproduction. Treatment of wild-type cells with rotenone, an inhibitor for complex I of the electron transport chain, increases the ROS levels. Likewise, treatment with mitoTEMPO, a SOD mimetic, rescues the ROS production in cells lacking vimentin. Together, these data show vimentin regulates neutrophil antimicrobial functions and alters ROS levels through regulation of mitochondrial activity.


Assuntos
Mitocôndrias , Neutrófilos , Espécies Reativas de Oxigênio , Vimentina , Espécies Reativas de Oxigênio/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Vimentina/metabolismo , Mitocôndrias/metabolismo , Animais , Camundongos , Inflamação/imunologia , Inflamação/metabolismo , Armadilhas Extracelulares/imunologia , Armadilhas Extracelulares/metabolismo , Fagocitose , Inflamassomos/metabolismo , Inflamassomos/imunologia , Citocinas/metabolismo , Humanos , Rotenona/farmacologia
11.
PLoS Pathog ; 20(8): e1012368, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39172744

RESUMO

The severity of COVID-19 is linked to excessive inflammation. Neutrophils represent a critical arm of the innate immune response and are major mediators of inflammation, but their role in COVID-19 pathophysiology remains poorly understood. We conducted transcriptomic profiling of neutrophils obtained from patients with mild and severe COVID-19, as well as from SARS-CoV-2 infected mice, in comparison to non-infected healthy controls. In addition, we investigated the inflammasome formation potential in neutrophils from patients and mice upon SARS-CoV-2 infection. Transcriptomic analysis of polymorphonuclear cells (PMNs), consisting mainly of mature neutrophils, revealed a striking type I interferon (IFN-I) gene signature in severe COVID-19 patients, contrasting with mild COVID-19 and healthy controls. Notably, low-density granulocytes (LDGs) from severe COVID-19 patients exhibited an immature neutrophil phenotype and lacked this IFN-I signature. Moreover, PMNs from severe COVID-19 patients showed heightened nigericin-induced caspase1 activation, but reduced responsiveness to exogenous inflammasome priming. Furthermore, IFN-I emerged as a priming stimulus for neutrophil inflammasomes. These findings suggest a potential role for neutrophil inflammasomes in driving inflammation during severe COVID-19. Altogether, these findings open promising avenues for targeted therapeutic interventions to mitigate the pathological processes associated with the disease.


Assuntos
COVID-19 , Inflamassomos , Interferon Tipo I , Neutrófilos , SARS-CoV-2 , COVID-19/imunologia , Humanos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Inflamassomos/imunologia , Inflamassomos/metabolismo , Animais , SARS-CoV-2/imunologia , Camundongos , Masculino , Feminino , Pessoa de Meia-Idade , Imunidade Inata , Adulto , Camundongos Endogâmicos C57BL
12.
Mol Immunol ; 173: 117-126, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39116800

RESUMO

Bacterial meningitis is a serious central nervous system (CNS) infection, claiming millions of human lives annually around the globe. The deadly infection involves severe inflammation of the protective sheath of the brain, i.e., meninges, and sometimes also consists of the brain tissue, called meningoencephalitis. Several inflammatory pathways involved in the pathogenesis of meningitis caused by Streptococcus pneumoniae, Neisseria meningitidis, Escherichia coli, Haemophilus influenzae, Mycobacterium tuberculosis, Streptococcus suis, etc. are mentioned in the scientific literature. Many in-vitro and in-vivo analyses have shown that after the disruption of the blood-brain barrier (BBB), these pathogens trigger several inflammatory pathways including Toll-Like Receptor (TLR) signaling in response to Pathogen-Associated Molecular Patterns (PAMPs), Nucleotide oligomerization domain (NOD)-like receptor-mediated signaling, pneumolysin related signaling, NF-κB signaling and many other pathways that lead to pro-inflammatory cascade and subsequent cytokine release including interleukine (IL)-1ß, tumor necrosis factor(TNF)-α, IL-6, IL-8, chemokine (C-X-C motif) ligand 1 (CXCL1) along with other mediators, leading to neuroinflammation. The activation of another protein complex, nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome, also takes place resulting in the maturation and release of IL-1ß and IL-18, hence potentiating neuroinflammation. This review aims to outline the inflammatory signaling pathways associated with the pathogenesis of bacterial meningitis leading to extensive pathological changes in neurons, astrocytes, oligodendrocytes, and other central nervous system cells.


Assuntos
Quimiocinas , Citocinas , Inflamação , Meningites Bacterianas , Transdução de Sinais , Humanos , Transdução de Sinais/imunologia , Meningites Bacterianas/imunologia , Meningites Bacterianas/microbiologia , Meningites Bacterianas/metabolismo , Citocinas/metabolismo , Citocinas/imunologia , Animais , Quimiocinas/metabolismo , Quimiocinas/imunologia , Inflamação/imunologia , Barreira Hematoencefálica/imunologia , Inflamassomos/imunologia , Inflamassomos/metabolismo
13.
J Innate Immun ; 16(1): 413-424, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39137733

RESUMO

BACKGROUND: The reemergence of monkeypox virus (Mpox, formerly monkeypox) in 2022 in non-endemic countries has raised significant concerns for global health due to its high transmissibility and mortality rate. A major challenge in combating Mpox is its ability to evade the host's innate immune system, the first line of defense against viral infections. SUMMARY: Mpox encodes various proteins that interfere with key antiviral pathways and mechanisms, such as the nuclear factor kappa B signaling, cytokine production, complement and inflammasome activation, and chemokine binding. These proteins modulate the expression and function of innate immune mediators, such as interferons, interleukins, and Toll-like receptors, and impair the recruitment and activation of innate immune cells, such as natural killer cells. By suppressing or altering these innate immune responses, Mpox enhances its replication and infection in the host tissues and organs, leading to systemic inflammation, tissue damage, and organ failure. KEY MESSAGES: This study reveals new insights into the molecular and cellular interactions between Mpox and the host's innate immune system. It identifies potential targets and strategies for antiviral interventions, highlighting the importance of understanding these interactions to develop effective treatments and improve global health responses to Mpox outbreaks.


Assuntos
Evasão da Resposta Imune , Imunidade Inata , Monkeypox virus , Mpox , Humanos , Animais , Monkeypox virus/imunologia , Mpox/imunologia , Transdução de Sinais , Inflamassomos/imunologia , Inflamassomos/metabolismo , Células Matadoras Naturais/imunologia , Citocinas/metabolismo , Citocinas/imunologia , NF-kappa B/metabolismo
14.
Int Immunopharmacol ; 141: 112891, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-39153310

RESUMO

In our investigation, we investigated the role of macrophage migration inhibitory factor (MIF), a key cytokine, in chronic nonbacterial prostatitis (CNP), an underexplored pathology. Elevated MIF expression was observed in the serum of individuals with chronic prostatitis-like symptoms (CP-LS) as well as in serum and tissue samples from experimental autoimmune prostatitis (EAP) mouse model. Treatment with ISO-1, a specific MIF antagonist, effectively mitigated prostatic inflammation and macrophage infiltration, thereby emphasizing the critical role of MIF in orchestrating immune responses within the prostate microenvironment. Further analyses revealed that MIF stimulates the PI3K/AKT and NLRP3 inflammasome pathways, which are integral to inflammation and cellular immunity. Pharmacological inhibition of the PI3K/AKT pathway by LY294002 substantially reduced prostatic inflammation and macrophage infiltration, potentially by inhibiting NLRP3 inflammasome activation. These findings collectively suggest that MIF is a potential diagnostic marker for CNP and suggest that targeting MIF or its downstream signalling pathways, PI3K/AKT and NLRP3, might represent a novel therapeutic strategy for this condition.


Assuntos
Doenças Autoimunes , Inflamassomos , Oxirredutases Intramoleculares , Fatores Inibidores da Migração de Macrófagos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fosfatidilinositol 3-Quinases , Prostatite , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Prostatite/imunologia , Prostatite/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Inflamassomos/metabolismo , Inflamassomos/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Humanos , Camundongos , Doenças Autoimunes/imunologia , Oxirredutases Intramoleculares/metabolismo , Oxirredutases Intramoleculares/antagonistas & inibidores , Modelos Animais de Doenças , Macrófagos/imunologia , Macrófagos/metabolismo , Adulto
15.
J Immunol Methods ; 533: 113744, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39147232

RESUMO

Inflammasomes play a major role in the immune response to infection, development of autoimmune disease, and control of cancer. Western blots were originally used in the early 2000s to characterize inflammasome activation. Since then, a panoply of techniques has been developed to characterize and visualize inflammasome activation in cells, tissues, and animals. This review article describes the most common techniques used by researchers in the inflammasome field and proposes that cell-specific characterization of inflammasome activation in tissues or animals may soon be commonly reported.


Assuntos
Inflamassomos , Inflamassomos/metabolismo , Inflamassomos/imunologia , Animais , Humanos
16.
J Exp Med ; 221(10)2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39212656

RESUMO

A growing number of patients presenting severe combined immunodeficiencies attributed to monoallelic RAC2 variants have been identified. The expression of the RHO GTPase RAC2 is restricted to the hematopoietic lineage. RAC2 variants have been described to cause immunodeficiencies associated with high frequency of infection, leukopenia, and autoinflammatory features. Here, we show that specific RAC2 activating mutations induce the NLRP3 inflammasome activation leading to the secretion of IL-1ß and IL-18 from macrophages. This activation depends on the activation state of the RAC2 variant and is mediated by the downstream kinase PAK1. Inhibiting the RAC2-PAK1-NLRP3 inflammasome pathway might be considered as a potential treatment for these patients.


Assuntos
Mutação com Ganho de Função , Inflamassomos , Interleucina-1beta , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteína RAC2 de Ligação ao GTP , Quinases Ativadas por p21 , Proteínas rac de Ligação ao GTP , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Inflamassomos/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Animais , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Camundongos , Interleucina-18/genética , Interleucina-18/metabolismo , Transdução de Sinais
17.
Front Immunol ; 15: 1447431, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39211040

RESUMO

Mammalian interleukin-22 (IL-22) attenuates organismal injury by inhibiting reactive oxygen species (ROS) and impeding the NLRP3 inflammasome activation. However, the role of fish IL-22 in this process remains unclear. We characterized MaIL-22, an IL-22 homolog in blunt snout bream (Megalobrama amblycephala). Despite its low sequence identity, it shares conserved structures and close evolutionary relationships with other teleost IL-22s. Furthermore, Aeromonas hydrophila (A. hydrophila) infection leads to tissue injury in M. amblycephala immune organs and concomitantly altered Mail-22 mRNA expression, suggesting that MaIL-22 was involved in the antimicrobial immune response. To explore MaIL-22's biological functions, we produced recombinant MaIL-22 (rMaIL-22) protein and demonstrated it significantly enhanced the survival of M. amblycephala post-A. hydrophila infection. To unravel its protective mechanisms, we explored the ROS/NLRP3 inflammasome axis and its downstream signaling responses. The results showed that rMaIL-22 treatment significantly elevated antioxidant enzyme (T-SOD, CAT and GSH-PX) activities to inhibit MDA activity and scavenge ROS in visceral tissues. Meanwhile, rMaIL-22 impeded the activation of NLRP3 inflammasome by suppressing NLRP3 protein and mRNA expression. This indicated that rMaIL-22 contributed to inhibit A. hydrophila-induced activation of the ROS/NLRP3 inflammasome axis. Consistent with these findings, rMaIL-22 treatment attenuated the expression of proinflammatory cytokines (il-1ß, tnf-α and il-6) and proapoptotic genes (caspase-3 and caspase-8) while promoting antiapoptotic genes (bcl-2b and mcl-1a) expression, ultimately mitigating tissue injury in visceral tissues. In conclusion, our research underscores MaIL-22's key role in microbial immune regulation, offering insights for developing IL-22-targeted therapies and breeding programs.


Assuntos
Aeromonas hydrophila , Apoptose , Doenças dos Peixes , Infecções por Bactérias Gram-Negativas , Inflamassomos , Inflamação , Interleucina 22 , Interleucinas , Proteína 3 que Contém Domínio de Pirina da Família NLR , Espécies Reativas de Oxigênio , Animais , Aeromonas hydrophila/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Inflamassomos/imunologia , Apoptose/efeitos dos fármacos , Infecções por Bactérias Gram-Negativas/imunologia , Espécies Reativas de Oxigênio/metabolismo , Interleucinas/metabolismo , Interleucinas/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Inflamação/imunologia , Proteínas de Peixes/metabolismo , Proteínas de Peixes/imunologia , Proteínas de Peixes/genética , Cyprinidae/imunologia , Cyprinidae/metabolismo , Transdução de Sinais
18.
Life Sci ; 354: 122951, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39127315

RESUMO

In the contemporary landscape of oncology, immunotherapy, represented by immune checkpoint blockade (ICB) therapy, stands out as a beacon of innovation in cancer treatment. Despite its promise, the therapy's progression is hindered by suboptimal clinical response rates. Addressing this challenge, the modulation of the NLRP3 inflammasome-GSDMD-mediated pyroptosis pathway holds promise as a means to augment the efficacy of immunotherapy. In the pathway, the NLRP3 inflammasome serves as a pivotal molecular sensor that responds to inflammatory stimuli within the organism. Its activation leads to the release of cytokines interleukin 1ß and interleukin 18 through the cleavage of GSDMD, thereby forming membrane pores and potentially resulting in pyroptosis. This cascade of processes exerts a profound impact on tumor development and progression, with its function and expression exhibiting variability across different tumor types and developmental stages. Consequently, understanding the specific roles of the NLRP3 inflammasome and GSDMD-mediated pyroptosis in diverse tumors is imperative for comprehending tumorigenesis and crafting precise therapeutic strategies. This review aims to elucidate the structure and activation mechanisms of the NLRP3 inflammasome, as well as the induction mechanisms of GSDMD-mediated pyroptosis. Additionally, we provide a comprehensive overview of the involvement of this pathway in various cancer types and its applications in tumor immunotherapy, nanotherapy, and other fields. Emphasis is placed on the feasibility of leveraging this approach to enhance ICB therapy within the field of immunotherapy. Furthermore, we discuss the potential applications of this pathway in other immunotherapy methods, such as chimeric antigen receptor T-cell (CAR-T) therapy and tumor vaccines.


Assuntos
Imunoterapia , Inflamassomos , Peptídeos e Proteínas de Sinalização Intracelular , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neoplasias , Proteínas de Ligação a Fosfato , Piroptose , Humanos , Piroptose/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Imunoterapia/métodos , Inflamassomos/metabolismo , Inflamassomos/imunologia , Proteínas de Ligação a Fosfato/metabolismo , Animais , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Gasderminas
19.
PLoS Pathog ; 20(8): e1012474, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39186780

RESUMO

The bacterium Vibrio vulnificus causes fatal septicemia in humans. Previously, we reported that an extracellular metalloprotease, vEP-45, secreted by V. vulnificus, undergoes self-proteolysis to generate a 34 kDa protease (vEP-34) by losing its C-terminal domain to produce the C-ter100 peptide. Moreover, we revealed that vEP-45 and vEP-34 proteases induce blood coagulation and activate the kallikrein/kinin system. However, the role of the C-ter100 peptide fragment released from vEP-45 in inducing inflammation is still unclear. Here, we elucidate, for the first time, the effects of C-ter100 on inducing inflammation and activating host innate immunity. Our results showed that C-ter100 could activate NF-κB by binding to the receptor TLR4, thereby promoting the secretion of inflammatory cytokines and molecules, such as TNF-α and nitric oxide (NO). Furthermore, C-ter100 could prime and activate the NLRP3 inflammasome (NLRP3, ASC, and caspase 1), causing IL-1ß secretion. In mice, C-ter100 induced the recruitment of immune cells, such as neutrophils and monocytes, along with histamine release into the plasma. Furthermore, the inflammatory response induced by C-ter100 could be effectively neutralized by an anti-C-ter100 monoclonal antibody (C-ter100Mab). These results demonstrate that C-ter100 can be a pathogen-associated molecular pattern (PAMP) that activates an innate immune response during Vibrio infection and could be a target for the development of antibiotics.


Assuntos
Imunidade Inata , Inflamação , Vibrio vulnificus , Animais , Camundongos , Inflamação/imunologia , Inflamação/metabolismo , Vibrio vulnificus/imunologia , Vibrioses/imunologia , Camundongos Endogâmicos C57BL , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/imunologia
20.
Arch Dermatol Res ; 316(8): 501, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39093484

RESUMO

The goal of this study is to investigate the impact of the rs35829419 SNP on the serum level of NLRP3, and to assess the relationship between NLRP3 and its SNP and vulnerability to Pityriasis versicolor. Pityriasis versicolor (PV) is one of the most frequent skin conditions linked to skin pigmentation changes. Malassezia plays a key role in the pathogenesis of PV. A case-control study, 50 patients with pityriasis versicolor and 44 healthy controls. Real-time PCR was used to genotype NLRP3 (rs35829419) and ELISA assay of NLRP3 levels in tissue samples. There was a significantly higher median NLPR3 levels in PV patients than controls. A significant predominance of A allele of Q 705 K was in patients than controls. The risk of having the disease in the presence of A allele is nearly 10 times than having C allele. In PV patients, there was a significant relationship between NLPR3 levels and Q 705 K genotypes with higher NLPR3 levels in AA genotype. A potential correlation between PV and the Q705K polymorphism, pointing to evidence of NLRP3 alteration in PV patients. The NLRP3 inflammasome may be an appropriate therapeutic target for Malassezia-associated skin disorders.


Assuntos
Genótipo , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Polimorfismo de Nucleotídeo Único , Pele , Tinha Versicolor , Humanos , Tinha Versicolor/diagnóstico , Tinha Versicolor/genética , Tinha Versicolor/microbiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Feminino , Masculino , Estudos de Casos e Controles , Adulto , Inflamassomos/genética , Inflamassomos/metabolismo , Inflamassomos/imunologia , Pele/patologia , Pele/microbiologia , Malassezia/isolamento & purificação , Malassezia/imunologia , Malassezia/genética , Adulto Jovem , Predisposição Genética para Doença , Pessoa de Meia-Idade , Alelos , Adolescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...