Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.538
Filtrer
1.
Balkan Med J ; 41(4): 286-297, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38966918

RÉSUMÉ

Background: Cannabidiol (CBD), extracted from Cannabis sativa, has anticancer, anti-inflammation, and analgesic effects. Nevertheless, its therapeutic effect and the mechanism by which it alleviates oral mucositis (OM) remain unclear. Aims: To explore the impact of CBD on OM in mice and on human oral keratinocyte (HOK) cells. Study Design: Expiremental study. Methods: The Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform, GeneCard, DisGeNET, and Gene Expression Omnibus databases were used to conduct therapeutic target gene screening for drugs against OM. Cytoscape software was used to build networks linking components, targets, and diseases. The STRING database facilitated analysis of intertarget action relationships, and the target genes were analyzed for Kyoto Encyclopedia of Genes and Genomes pathway enrichment. Occurrence of serum inflammation-related factors, hematoxylin and eosin staining, and immunohistochemistry were used to assess OM injury. Cell proliferation, migration, pyroptosis, and apoptosis of HOK cells under different treatments were assessed. Molecular mechanisms were elucidated through western blot and quantitative real-time polymerase chain reaction analyses. Results: A total of 49 overlapping genes were pinpointed as potential targets, with NF-κB1, PIK3R1, NF-κBIA, and AKT1 being recognized as hub genes among them. Additionally, the PI3K/Akt/NF-κB and interleukin-17 signaling pathways were identified as relevant. Our in vivo experiments showed that CBD significantly reduced the proportion of lesion area, mitigated oral mucosal tissue lesions, and downregulated the expression levels of genes and levels of proteins, including NLRP3, P65, AKT, and PI3K. In vitro experiments indicated that CBD enhanced HOK cell proliferation and migration and reduced apoptosis through inhibition of the PI3K/Akt/NF-κB signaling pathway and pyroptosis. Conclusion: Our findings suggest a novel mechanism for controlling OM, in which CBD suppresses the PI3K/Akt/NF-κB signaling pathway and pyroptosis, thereby mitigating OM symptoms.


Sujet(s)
Cannabidiol , Facteur de transcription NF-kappa B , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Pyroptose , Stomatite , Cannabidiol/pharmacologie , Cannabidiol/usage thérapeutique , Animaux , Pyroptose/effets des médicaments et des substances chimiques , Souris , Stomatite/traitement médicamenteux , Facteur de transcription NF-kappa B/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/analyse , Phosphatidylinositol 3-kinases/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
2.
Int J Chron Obstruct Pulmon Dis ; 19: 1491-1513, 2024.
Article de Anglais | MEDLINE | ID: mdl-38957709

RÉSUMÉ

Background: Chronic obstructive pulmonary disease (COPD) stands as a predominant cause of global morbidity and mortality. This study aims to elucidate the relationship between pyroptosis-related genes (PRGs) and COPD diagnosis in the context of immune infiltration, ultimately proposing a PRG-based diagnostic model for predicting COPD outcomes. Methods: Clinical data and PRGs of COPD patients were sourced from the GEO database. The "ConsensusClusterPlus" package was employed to generate molecular subtypes derived from PRGs that were identified through differential expression analysis and LASSO Cox analysis. A diagnostic signature including eight genes (CASP4, CASP5, ELANE, GPX4, NLRP1, GSDME, NOD1and IL18) was also constructed. Immune cell infiltration calculated by the ESTIMATE score, Stroma scores and Immune scores were also compared on the basis of pyroptosis-related molecular subtypes and the risk signature. We finally used qRT - PCR to detect the expression levels of eight genes in COPD patient and normal. Results: The diagnostic model, anchored on eight PRGs, underwent validation with an independent experimental cohort. The area under the receiver operating characteristic (ROC) curves (AUC) for the diagnostic model showcased values of 0.809, 0.765, and 0.956 for the GSE76925, GSE8545, and GSE5058 datasets, respectively. Distinct expression patterns and clinical attributes of PRGs were observed between the comparative groups, with functional analysis underscoring a disparity in immune-related functions between them. Conclusion: In this study, we developed a potential as diagnostic biomarkers for COPD and have a significant role in modulating the immune response. Such insights pave the way for novel diagnostic and therapeutic strategies for COPD.


Sujet(s)
Bases de données génétiques , Valeur prédictive des tests , Broncho-pneumopathie chronique obstructive , Pyroptose , Humains , Broncho-pneumopathie chronique obstructive/génétique , Broncho-pneumopathie chronique obstructive/diagnostic , Broncho-pneumopathie chronique obstructive/immunologie , Pyroptose/génétique , Analyse de profil d'expression de gènes , Poumon/immunologie , Mâle , Femelle , Adulte d'âge moyen , Marqueurs génétiques , Études cas-témoins , Transcriptome , Sujet âgé , Reproductibilité des résultats , Prédisposition génétique à une maladie , Pronostic
3.
Cell Death Dis ; 15(7): 479, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965211

RÉSUMÉ

TLR4 and TNFR1 signalling promotes potent proinflammatory signal transduction events, thus, are often hijacked by pathogenic microorganisms. We recently reported that myeloid cells retaliate Yersinia blockade of TAK1/IKK signalling by triggering RIPK1-dependent caspase-8 activation that promotes downstream GSDMD and GSDME-mediated pyroptosis in macrophages and neutrophils respectively. However, the upstream signalling events for RIPK1 activation in these cells are not well defined. Here, we demonstrate that unlike in macrophages, RIPK1-driven pyroptosis and cytokine priming in neutrophils are driven through TNFR1 signalling, while TLR4-TRIF signalling is dispensable. Furthermore, we demonstrate that activation of RIPK1-dependent pyroptosis in neutrophils during Yersinia infection requires IFN-γ priming, which serves to induce surface TNFR1 expression and amplify soluble TNF secretion. In contrast, macrophages utilise both TNFR1 and TLR4-TRIF signalling to trigger cell death, but only require TRIF but not autocrine TNFR1 for cytokine production. Together, these data highlight the emerging theme of cell type-specific regulation in cell death and immune signalling in myeloid cells.


Sujet(s)
Macrophages , Granulocytes neutrophiles , Pyroptose , Receptor-Interacting Protein Serine-Threonine Kinases , Récepteur au facteur de nécrose tumorale de type I , Transduction du signal , Récepteur de type Toll-4 , Macrophages/métabolisme , Granulocytes neutrophiles/métabolisme , Animaux , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Récepteur au facteur de nécrose tumorale de type I/métabolisme , Souris , Récepteur de type Toll-4/métabolisme , Protéines adaptatrices du transport vésiculaire/métabolisme , Souris de lignée C57BL , Interféron gamma/métabolisme , Souris knockout
4.
Cancer Immunol Immunother ; 73(9): 177, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954046

RÉSUMÉ

Paclitaxel and anthracycline-based chemotherapy is one of the standard treatment options for breast cancer. However, only about 6-30% of breast cancer patients achieved a pathological complete response (pCR), and the mechanism responsible for the difference is still unclear. In this study, random forest algorithm was used to screen feature genes, and artificial neural network (ANN) algorithm was used to construct an ANN model for predicting the efficacy of neoadjuvant chemotherapy for breast cancer. Furthermore, digital pathology, cytology, and molecular biology experiments were used to verify the relationship between the efficacy of neoadjuvant chemotherapy and immune ecology. It was found that paclitaxel and doxorubicin, an anthracycline, could induce typical pyroptosis and bubbling in breast cancer cells, accompanied by gasdermin E (GSDME) cleavage. Paclitaxel with LDH release and Annexin V/PI doubule positive cell populations, and accompanied by the increased release of damage-associated molecular patterns, HMGB1 and ATP. Cell coculture experiments also demonstrated enhanced phagocytosis of macrophages and increased the levels of IFN-γ and IL-2 secretion after paclitaxel treatment. Mechanistically, GSDME may mediate paclitaxel and doxorubicin-induced pyroptosis in breast cancer cells through the caspase-9/caspase-3 pathway, activate anti-tumor immunity, and promote the efficacy of paclitaxel and anthracycline-based neoadjuvant chemotherapy. This study has practical guiding significance for the precision treatment of breast cancer, and can also provide ideas for understanding molecular mechanisms related to the chemotherapy sensitivity.


Sujet(s)
Tumeurs du sein , Traitement néoadjuvant , Pyroptose , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/immunologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Humains , Pyroptose/effets des médicaments et des substances chimiques , Femelle , Traitement néoadjuvant/méthodes , Souris , Animaux , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Gasdermines
5.
Stem Cell Res Ther ; 15(1): 194, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956719

RÉSUMÉ

BACKGROUND: Repairation of bone defects remains a major clinical problem. Constructing bone tissue engineering containing growth factors, stem cells, and material scaffolds to repair bone defects has recently become a hot research topic. Nerve growth factor (NGF) can promote osteogenesis of bone marrow mesenchymal stem cells (BMSCs), but the low survival rate of the BMSCs during transplantation remains an unresolved issue. In this study, we investigated the therapeutic effect of BMSCs overexpression of NGF on bone defect by inhibiting pyroptosis. METHODS: The relationship between the low survival rate and pyroptosis of BMSCs overexpressing NGF in localized inflammation of fractures was explored by detecting pyroptosis protein levels. Then, the NGF+/BMSCs-NSA-Sca bone tissue engineering was constructed by seeding BMSCs overexpressing NGF on the allograft bone scaffold and adding the pyroptosis inhibitor necrosulfonamide(NSA). The femoral condylar defect model in the Sprague-Dawley (SD) rat was studied by micro-CT, histological, WB and PCR analyses in vitro and in vivo to evaluate the regenerative effect of bone repair. RESULTS: The pyroptosis that occurs in BMSCs overexpressing NGF is associated with the nerve growth factor receptor (P75NTR) during osteogenic differentiation. Furthermore, NSA can block pyroptosis in BMSCs overexpression NGF. Notably, the analyses using the critical-size femoral condylar defect model indicated that the NGF+/BMSCs-NSA-Sca group inhibited pyroptosis significantly and had higher osteogenesis in defects. CONCLUSION: NGF+/BMSCs-NSA had strong osteogenic properties in repairing bone defects. Moreover, NGF+/BMSCs-NSA-Sca mixture developed in this study opens new horizons for developing novel tissue engineering constructs.


Sujet(s)
Cellules souches mésenchymateuses , Facteur de croissance nerveuse , Ostéogenèse , Rat Sprague-Dawley , Structures d'échafaudage tissulaires , Animaux , Facteur de croissance nerveuse/métabolisme , Facteur de croissance nerveuse/génétique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Rats , Structures d'échafaudage tissulaires/composition chimique , Régénération osseuse , Allogreffes , Mâle , Ingénierie tissulaire/méthodes , Pyroptose , Sulfonamides/pharmacologie , Différenciation cellulaire , Transplantation de cellules souches mésenchymateuses/méthodes , Transplantation osseuse/méthodes
6.
Proc Natl Acad Sci U S A ; 121(29): e2400883121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38980908

RÉSUMÉ

Gasdermin D (GSDMD)-mediated pyroptotic cell death drives inflammatory cytokine release and downstream immune responses upon inflammasome activation, which play important roles in host defense and inflammatory disorders. Upon activation by proteases, the GSDMD N-terminal domain (NTD) undergoes oligomerization and membrane translocation in the presence of lipids to assemble pores. Despite intensive studies, the molecular events underlying the transition of GSDMD from an autoinhibited soluble form to an oligomeric pore form inserted into the membrane remain incompletely understood. Previous work characterized S-palmitoylation for gasdermins from bacteria, fungi, invertebrates, as well as mammalian gasdermin E (GSDME). Here, we report that a conserved residue Cys191 in human GSDMD was S-palmitoylated, which promoted GSDMD-mediated pyroptosis and cytokine release. Mutation of Cys191 or treatment with palmitoyltransferase inhibitors cyano-myracrylamide (CMA) or 2-bromopalmitate (2BP) suppressed GSDMD palmitoylation, its localization to the membrane and dampened pyroptosis or IL-1ß secretion. Furthermore, Gsdmd-dependent inflammatory responses were alleviated by inhibition of palmitoylation in vivo. By contrast, coexpression of GSDMD with palmitoyltransferases enhanced pyroptotic cell death, while introduction of exogenous palmitoylation sequences fully restored pyroptotic activities to the C191A mutant, suggesting that palmitoylation-mediated membrane localization may be distinct from other molecular events such as GSDMD conformational change during pore assembly. Collectively, our study suggests that S-palmitoylation may be a shared regulatory mechanism for GSDMD and other gasdermins, which points to potential avenues for therapeutically targeting S-palmitoylation of gasdermins in inflammatory disorders.


Sujet(s)
Cystéine , Protéines et peptides de signalisation intracellulaire , Lipoylation , Protéines de liaison aux phosphates , Pyroptose , Protéines de liaison aux phosphates/métabolisme , Protéines de liaison aux phosphates/génétique , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Cystéine/métabolisme , Animaux , Souris , Cytokines/métabolisme , Cellules HEK293 , Inflammasomes/métabolisme , Gasdermines
7.
Immun Inflamm Dis ; 12(7): e1303, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967379

RÉSUMÉ

BACKGROUND: Psoriasis refers to a highly prevalent and immunologically mediated dermatosis with considerable deterioration in life quality. Wogonin, a sort of flavonoid, has been mentioned to elicit protective activities in skin diseases. However, whether Wogonin is implicated in the treatment of psoriasis and its specific mechanisms are not fully understood. AIM: The present work attempted to elaborate the role of Wogonin during the process of psoriasis and to concentrate on the associated action mechanism. METHODS: Cell counting kit-8 (CCK-8) method was initially applied to assay the viability of human keratinocyte HaCaT cells treated by varying concentrations of Wogonin. To mimic psoriasis in vitro, HaCaT cells were exposed to M5 cytokines. CCK-8 and 5-Ethynyl-2'-deoxyuridine  assays were adopted for the measurement of cell proliferation. Inflammatory levels were examined with enzyme-linked immunosorbent assay. Immunofluorescence staining tested nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) and Caspase-1 expressions. Western blot examined the protein expressions of proliferation-, inflammation-, pyroptosis-associated factors, and NLRP3. RESULTS: Wogonin treatment antagonized the proliferation, inflammatory response, and NLRP3/caspase-1/Gasdermin-D (GSDMD)-mediated pyroptosis in M5-challenged HaCaT cells. Besides, NLRP3 elevation partially abrogated the effects of Wogonin on M5-induced proliferation, inflammatory response, and NLRP3/caspase-1/GSDMD-mediated pyroptosis in HaCaT cells. CONCLUSION: In a word, Wogonin might exert anti-proliferation, anti-inflammatory and anti-pyroptosis activities in M5-induced cell model of psoriasis and the blockade of NLRP3/Caspase-1/GSDMD pathway might be recognized as a potential mechanism underlying the protective mechanism of Wogonin in psoriasis, suggesting Wogonin as a prospective anti-psoriasis drug.


Sujet(s)
Caspase-1 , Prolifération cellulaire , Flavanones , Kératinocytes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Transduction du signal , Humains , Flavanones/pharmacologie , Pyroptose/effets des médicaments et des substances chimiques , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Caspase-1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intracellulaire/métabolisme , Psoriasis/traitement médicamenteux , Psoriasis/métabolisme , Psoriasis/anatomopathologie , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Cellules HaCaT , Lignée cellulaire , Gasdermines , Protéines de liaison aux phosphates
8.
Sci Immunol ; 9(97): eadn0178, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996010

RÉSUMÉ

Virus-induced cell death is a key contributor to COVID-19 pathology. Cell death induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is well studied in myeloid cells but less in its primary host cell type, angiotensin-converting enzyme 2 (ACE2)-expressing human airway epithelia (HAE). SARS-CoV-2 induces apoptosis, necroptosis, and pyroptosis in HAE organotypic cultures. Single-cell and limiting-dilution analysis revealed that necroptosis is the primary cell death event in infected cells, whereas uninfected bystanders undergo apoptosis, and pyroptosis occurs later during infection. Mechanistically, necroptosis is induced by viral Z-RNA binding to Z-DNA-binding protein 1 (ZBP1) in HAE and lung tissues from patients with COVID-19. The Delta (B.1.617.2) variant, which causes more severe disease than Omicron (B1.1.529) in humans, is associated with orders of magnitude-greater Z-RNA/ZBP1 interactions, necroptosis, and disease severity in animal models. Thus, Delta induces robust ZBP1-mediated necroptosis and more disease severity.


Sujet(s)
COVID-19 , Nécroptose , Pyroptose , Protéines de liaison à l'ARN , Muqueuse respiratoire , SARS-CoV-2 , Humains , SARS-CoV-2/immunologie , COVID-19/immunologie , COVID-19/anatomopathologie , Nécroptose/immunologie , Animaux , Muqueuse respiratoire/virologie , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/anatomopathologie , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris , Mort cellulaire/immunologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Apoptose/immunologie
9.
Sci Immunol ; 9(97): eadp8170, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996011

RÉSUMÉ

Upon SARS-CoV-2 infection, infected cells undergo necroptosis, whereas delayed apoptosis and pyroptosis occur in uninfected, bystander cells, thus providing a plausible explanation for the extensive injury among myriad uninfected cells.


Sujet(s)
COVID-19 , Nécroptose , Pyroptose , SARS-CoV-2 , Humains , COVID-19/immunologie , COVID-19/anatomopathologie , SARS-CoV-2/immunologie , Pyroptose/immunologie , Nécroptose/immunologie , Apoptose/immunologie , Mort cellulaire/immunologie , Animaux
10.
J Nanobiotechnology ; 22(1): 403, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982427

RÉSUMÉ

BACKGROUND: Following spinal cord injury (SCI), the inflammatory storm initiated by microglia/macrophages poses a significant impediment to the recovery process. Exosomes play a crucial role in the transport of miRNAs, facilitating essential cellular communication through the transfer of genetic material. However, the miRNAs from iPSC-NSCs-Exos and their potential mechanisms leading to repair after SCI remain unclear. This study aims to explore the role of iPSC-NSCs-Exos in microglia/macrophage pyroptosis and reveal their potential mechanisms. METHODS: iPSC-NSCs-Exos were characterized and identified using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot. A mouse SCI model and a series of in vivo and in vitro experiments were conducted to investigate the therapeutic effects of iPSC-NSCs-Exos. Subsequently, miRNA microarray analysis and rescue experiments were performed to confirm the role of miRNAs in iPSC-NSCs-Exos in SCI. Mechanistic studies were carried out using Western blot, luciferase activity assays, and RNA-ChIP. RESULTS: Our findings revealed that iPSC-NSCs-derived exosomes inhibited microglia/macrophage pyroptosis at 7 days post-SCI, maintaining myelin integrity and promoting axonal growth, ultimately improving mice motor function. The miRNA microarray showed let-7b-5p to be highly enriched in iPSC-NSCs-Exos, and LRIG3 was identified as the target gene of let-7b-5p. Through a series of rescue experiments, we uncovered the connection between iPSC-NSCs and microglia/macrophages, revealing a novel target for treating SCI. CONCLUSION: In conclusion, we discovered that iPSC-NSCs-derived exosomes can package and deliver let-7b-5p, regulating the expression of LRIG3 to ameliorate microglia/macrophage pyroptosis and enhance motor function in mice after SCI. This highlights the potential of combined therapy with iPSC-NSCs-Exos and let-7b-5p in promoting functional recovery and limiting inflammation following SCI.


Sujet(s)
Exosomes , Cellules souches pluripotentes induites , Macrophages , microARN , Microglie , Pyroptose , Traumatismes de la moelle épinière , Animaux , Exosomes/métabolisme , microARN/génétique , microARN/métabolisme , Traumatismes de la moelle épinière/thérapie , Traumatismes de la moelle épinière/métabolisme , Cellules souches pluripotentes induites/métabolisme , Souris , Microglie/métabolisme , Macrophages/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine , Femelle , Mâle
11.
World J Gastroenterol ; 30(23): 2931-2933, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38946877

RÉSUMÉ

In this editorial we comment on the article published in a recent issue of the World Journal of Gastroenterology. Acute liver failure (ALF) is a critical condition characterized by rapid hepatocellular injury and organ dysfunction, and it often necessitates liver transplant to ensure patient survival. Recent research has elucidated the involvement of distinct cell death pathways, namely ferroptosis and pyroptosis, in the pathogenesis of ALF. Ferroptosis is driven by iron-dependent lipid peroxidation, whereas pyroptosis is an inflammatory form of cell death; both pathways contribute to hepatocyte death and exacerbate tissue damage. This comprehensive review explores the interplay between ferroptosis and pyroptosis in ALF, highlighting the role of key regulators such as silent information regulator sirtuin 1. Insights from clinical and preclinical studies provide valuable perspectives on the dysregulation of cell death pathways in ALF and the therapeutic potential of targeting these pathways. Collaboration across multiple disciplines is essential for translating the experimental insights into effective treatments for this life-threatening condition.


Sujet(s)
Ferroptose , Défaillance hépatique aigüe , Pyroptose , Animaux , Humains , Hépatocytes/métabolisme , Fer/métabolisme , Peroxydation lipidique , Foie/métabolisme , Foie/anatomopathologie , Défaillance hépatique aigüe/métabolisme , Défaillance hépatique aigüe/thérapie , Transplantation hépatique , Transduction du signal , Sirtuine-1/métabolisme
12.
Front Immunol ; 15: 1381778, 2024.
Article de Anglais | MEDLINE | ID: mdl-38947336

RÉSUMÉ

Background: The interaction between pyroptosis-a form of programmed cell death-and tumor immunity represents a burgeoning field of interest. Pyroptosis exhibits a dual role in cancer: it can both promote tumor development and counteract it by activating immune responses that inhibit tumor evasion and encourage cell death. Current tumor immunotherapy strategies, notably CAR-T cell therapy and immune checkpoint inhibitors (ICIs), alongside the potential of certain traditional Chinese medicinal compounds, highlight the intricate relationship between pyroptosis and cancer immunity. As research delves deeper into pyroptosis mechanisms within tumor therapy, its application in enhancing tumor immune responses emerges as a novel research avenue. Purpose: This review aims to elucidate the mechanisms underlying pyroptosis, its impact on tumor biology, and the advancements in tumor immunotherapy research. Methods: A comprehensive literature review was conducted across PubMed, Embase, CNKI, and Wanfang Database from the inception of the study until August 22, 2023. The search employed keywords such as "pyroptosis", "cancer", "tumor", "mechanism", "immunity", "gasdermin", "ICB", "CAR-T", "PD-1", "PD-L1", "herbal medicine", "botanical medicine", "Chinese medicine", "traditional Chinese medicine", "immunotherapy", linked by AND/OR, to capture the latest findings in pyroptosis and tumor immunotherapy. Results: Pyroptosis is governed by a complex mechanism, with the Gasdermin family playing a pivotal role. While promising for tumor immunotherapy application, research into pyroptosis's effect on tumor immunity is still evolving. Notably, certain traditional Chinese medicine ingredients have been identified as potential pyroptosis inducers, meriting further exploration. Conclusion: This review consolidates current knowledge on pyroptosis's role in tumor immunotherapy. It reveals pyroptosis as a beneficial factor in the immunotherapeutic landscape, suggesting that leveraging pyroptosis for developing novel cancer treatment strategies, including those involving traditional Chinese medicine, represents a forward-looking approach in oncology.


Sujet(s)
Immunothérapie , Tumeurs , Pyroptose , Pyroptose/immunologie , Pyroptose/effets des médicaments et des substances chimiques , Humains , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Immunothérapie/méthodes , Animaux , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques
13.
Cell Mol Life Sci ; 81(1): 295, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38977508

RÉSUMÉ

Nod-like receptor family pyrin-containing protein 3 (NLRP3) inflammasome plays a pathologic role in metabolic dysfunction-associated steatohepatitis (MASH), but the molecular mechanism regulating the NLRP3 inflammasome activation in hepatocellular lipotoxicity remains largely unknown. Bromodomain-containing protein 4 (BRD4) has emerged as a key epigenetic reader of acetylated lysine residues in enhancer regions that control the transcription of key genes. The aim of this study is to investigate if and how BRD4 regulated the NLRP3 inflammasome activation and pyroptosis in MASH. Using the AML12 and primary mouse hepatocytes stimulated by palmitic acid (PA) as an in vitro model of hepatocellular lipotoxicity, we found that targeting BRD4 by genetic knockdown or a selective BRD4 inhibitor MS417 protected against hepatosteatosis; and this protective effect was attributed to inhibiting the activation of NLRP3 inflammasome and reducing the expression of Caspase-1, gasdermin D (GSDMD), interleukin (IL)-1ß and IL-6. Moreover, BRD4 inhibition limited the voltage-dependent anion channel-1 (VDAC1) expression and oligomerization in PA-treated AML12 hepatocytes, thereby suppressing the NLRP3 inflammasome activation. Additionally, the expression of BRD4 enhanced in MASH livers of humans. Mechanistically, BRD4 was upregulated during hepatocellular lipotoxicity that in turn modulated the active epigenetic mark H3K27ac at the promoter regions of the Vdac and Gsdmd genes, thereby enhancing the expression of VDAC and GSDMD. Altogether, our data provide novel insights into epigenetic mechanisms underlying BRD4 activating the NLRP3 inflammasome and promoting GSDMD-mediated pyroptosis in hepatocellular lipotoxicity. Thus, BRD4 might serve as a novel therapeutic target for the treatment of MASH.


Sujet(s)
Hépatocytes , Inflammasomes , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéines de liaison aux phosphates , Pyroptose , Facteurs de transcription , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/anatomopathologie , Pyroptose/effets des médicaments et des substances chimiques , Protéines de liaison aux phosphates/métabolisme , Protéines de liaison aux phosphates/génétique , Inflammasomes/métabolisme , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Acide palmitique/pharmacologie , Mâle , Indènes/pharmacologie , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Sulfonamides/pharmacologie , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Protéines du cycle cellulaire , Furanes , Gasdermines , Protéines contenant un bromodomaine , Protéines nucléaires
14.
Clin Exp Med ; 24(1): 145, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38960987

RÉSUMÉ

Pyroptosis-related long-noncoding RNAs (PRlncRNAs) play an important role in cancer progression. However, their role in lung squamous cell carcinoma (LUSC) is unclear. A risk model was constructed using the least absolute shrinkage and selection operator (LASSO) Cox regression analysis based on RNA sequencing data from The Cancer Genome Atlas database. The LUSC cohort was divided into high- and low-risk groups based on the median risk score. For the prognostic value of the model, the Kaplan-Meier analysis, log-rank test, and Cox regression analysis were performed. A nomogram was constructed to predict the prognosis of patients, using a risk score and clinical parameters such as age, sex, clinical stage, and tumor node metastasis classification (TNM) stage. Afterwards, six common algorithms were employed to assess the invasion of immune cells. The Gene Set Enrichment Analysis (GSEA) was conducted to identify differences between patients at high and low risk. Furthermore, the pRRophetic package was employed to forecast the half-maximal inhibitory doses of prevalent chemotherapeutic drugs, while the tumor immune dysfunction and exclusion score was computed to anticipate the response to immunotherapy. The expression levels of the seven PRlncRNAs were examined in both LUSC and normal lung epithelial cell lines using RT-qPCR. Proliferation, migration, and invasion assays were also carried out to investigate the role of MIR193BHG in LUSC cells. Patients in the low-risk group showed prolonged survival in the total cohort or subgroup analysis. The Cox regression analysis showed that the risk model could act as an independent prognostic factor for patients with LUSC. The results of GSEA analysis revealed that the high-risk group showed enrichment of cytokine pathways, Janus tyrosine kinase/signal transducer and activator of the transcription signalling pathway, and Toll-like receptor pathway. Conversely, the low-risk group showed enrichment of several gene repair pathways. Furthermore, the risk score was positively correlated with immune cell infiltration. Moreover, patients in the high-risk category showed reduced responsiveness to conventional chemotherapeutic medications and immunotherapy. The majority of the long noncoding RNAs in the risk model were confirmed to be overexpressed in LUSC cell lines compared to normal lung epithelial cell lines by in vitro tests. Further studies have shown that downregulating the expression of MIR193BHG may inhibit the growth, movement, and infiltration capabilities of LUSC cells, whereas increasing the expression of MIR193BHG could enhance these malignant tendencies. This study found that PRlncRNAs were linked to the prognosis of LUSC patients. The risk model, evaluated across various clinical parameters and treatment modalities, shows potential as a future reference for clinical applications.


Sujet(s)
Carcinome épidermoïde , Tumeurs du poumon , Pyroptose , ARN long non codant , Humains , ARN long non codant/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/thérapie , Tumeurs du poumon/mortalité , Mâle , Femelle , Carcinome épidermoïde/génétique , Carcinome épidermoïde/thérapie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/mortalité , Pronostic , Pyroptose/génétique , Immunothérapie , Adulte d'âge moyen , Nomogrammes , Régulation de l'expression des gènes tumoraux , Marqueurs biologiques tumoraux/génétique , Sujet âgé , Lignée cellulaire tumorale
15.
Hepatol Commun ; 8(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967580

RÉSUMÉ

BACKGROUND: lNUAK1 is strongly associated with organ fibrosis, but its causal mechanism for modulating lipid metabolism and hepatic inflammation underlying MASH has not been fully clarified. METHOD: In our study, human liver tissues from patients with MASH and control subjects were obtained to evaluate NUAK1 expression. MASH models were established using C57BL/6 mice. Liver damage and molecular mechanisms of the NUAK1-Caspase 6 signaling were tested in vivo and in vitro. RESULTS: In the clinical arm, NUAK1 expression was upregulated in liver samples from patients with MASH. Moreover, increased NUAK1 was detected in mouse MASH models. NUAK1 inhibition ameliorated steatohepatitis development in MASH mice accompanied by the downregulation of hepatic steatosis and fibrosis. Intriguingly, NUAK1 was found to facilitate Caspase 6 activation and trigger pyroptosis in MASH-stressed livers. Disruption of hepatocytes Caspase 6 decreased MASH-induced liver inflammation with upregulated TAK1 but diminished RIPK1. Moreover, we found that NUAK1/Caspase 6 axis inhibition could accelerate the interaction between TAK1 and RIPK1, which in turn led to the degradation of RIPK1. CONCLUSIONS: In summary, our study elucidates that NUAK1-Caspase 6 signaling controls inflammation activation in MASH through the interaction between TAK1 and RIPK1, which is crucial for controlling pyroptosis and promoting the progression of MASH.


Sujet(s)
Caspase-6 , Évolution de la maladie , Souris de lignée C57BL , Pyroptose , Animaux , Souris , Humains , Caspase-6/métabolisme , Mâle , Modèles animaux de maladie humaine , Transduction du signal , Stéatose hépatique/anatomopathologie , Stéatose hépatique/métabolisme , Inflammation/métabolisme , Foie/anatomopathologie , Foie/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , MAP Kinase Kinase Kinases/métabolisme
16.
Sci Rep ; 14(1): 15564, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38971897

RÉSUMÉ

Aortic dissection (AD) is a life-threatening condition with a high mortality rate and without effective pharmacological therapies. Our previous study illustrated that leukocyte immunoglobulin-like receptor B4 (LILRB4) knockdown promoted the contractile phenotypic switch and apoptosis of AD cells. This study aimed to further investigate the role of LILRB4 in animal models of AD and elucidate its underlying molecular mechanisms. Animal models of AD were established using 0.1% beta-aminopropionitrile and angiotensin II and an in vitro model was developed using platelet-derived growth factor BB (PDGF-BB). The effects of LILRB4 knockdown on histopathological changes, pyroptosis, phenotype transition, extracellular matrix (ECM), and Janus kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3) pathways were assessed using a series of in vivo and in vitro assays. The effects of the JAK2 inhibitor AG490 on AD cell function, phenotypic transition, and ECM were explored. LILRB4 was highly expressed in AD and its knockdown increased survival rate, reduced AD incidence, and alleviated histopathological changes in the AD mouse model. Furthermore, LILRB4 knockdown promoted contractile phenotype switch, stabilized the ECM, and inhibited pyroptosis. Mechanistically, LILRB4 knockdown inhibited the JAK2/STAT3 signaling pathway. JAK2 inhibitor AG490 inhibited cell viability and migration, enhanced apoptosis, induced G0/G1 cell cycle arrest, and suppressed S-phase progression in PDGF-BB-stimulated human aortic smooth muscle cells. LILRB4 knockdown suppresses AD development by inhibiting pyroptosis and the JAK2/STAT3 signaling pathway.


Sujet(s)
, Modèles animaux de maladie humaine , Kinase Janus-2 , Pyroptose , Facteur de transcription STAT-3 , Transduction du signal , Kinase Janus-2/métabolisme , Kinase Janus-2/génétique , /métabolisme , /anatomopathologie , /génétique , Animaux , Facteur de transcription STAT-3/métabolisme , Pyroptose/génétique , Souris , Humains , Techniques de knock-down de gènes , Mâle , Souris de lignée C57BL , Tyrphostines/pharmacologie
17.
Nat Commun ; 15(1): 5730, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977695

RÉSUMÉ

The circular RNA (circRNA) family is a group of endogenous non-coding RNAs (ncRNAs) that have critical functions in multiple physiological and pathological processes, including inflammation, cancer, and cardiovascular diseases. However, their roles in regulating innate immune responses remain unclear. Here, we define Cell division cycle 42 (CDC42)-165aa, a protein encoded by circRNA circCDC42, which is overexpressed in Klebsiella pneumoniae (KP)-infected alveolar macrophages. High levels of CDC42-165aa induces the hyperactivation of Pyrin inflammasomes and aggravates alveolar macrophage pyroptosis, while the inhibition of CDC42-165aa reduces lung injury in mice after KP infection by inhibiting Pyrin inflammasome-mediated pyroptosis. Overall, these results demonstrate that CDC42-165aa stimulates Pyrin inflammasome by inhibiting CDC42 GTPase activation and provides a potential clinical target for pathogenic bacterial infection in clinical practice.


Sujet(s)
Inflammasomes , Infections à Klebsiella , Klebsiella pneumoniae , Souris de lignée C57BL , Pyroptose , Protéine G cdc42 , Animaux , Pyroptose/génétique , Infections à Klebsiella/immunologie , Infections à Klebsiella/microbiologie , Infections à Klebsiella/métabolisme , Souris , Inflammasomes/métabolisme , Protéine G cdc42/métabolisme , Protéine G cdc42/génétique , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/microbiologie , Mâle , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Humains , Immunité innée , Macrophages/métabolisme , Macrophages/immunologie , Macrophages/microbiologie , Protéines adaptatrices de signalisation CARD
18.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000237

RÉSUMÉ

Pyroptosis, known as one typical mode of programmed cell death, is generally characterized by the cleaved gasdermin family (GSDMs) forming pores in the cell membrane and inducing cell rupture, and the activation of aspartate-specific proteases (caspases) has also been found during this process. Diabetic Kidney Disease (DKD) is caused by the complication of diabetes in the kidney, and the most important kidney's function, Glomerular Filtration Rate (GFR), happens to drop to less than 90% of its usual and even lead to kidney failure in severe cases. The persistent inflammatory state induced by high blood glucose implies the key pathology of DKD, and growing evidence shows that pyroptosis serves as a significant contributor to this chronic immune-mediated inflammatory disorder. Currently, the expanded discovery of GSDMs, pyroptosis, and its association with innate immunity has been more attractive, and overwhelming research is needed to sort out the implication of pyroptosis in DKD pathology. In this review, we comb both classical studies and newly founds on pyroptosis, prick off the novel awakening of pyroptosis in DKD, and center on the significance of pyroptosis in DKD treatment, aiming to provide new research targets and treatment strategies on DKD.


Sujet(s)
Néphropathies diabétiques , Pyroptose , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Humains , Animaux , Immunité innée
19.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-39000412

RÉSUMÉ

Biological aging results from an accumulation of damage in the face of reduced resilience. One major driver of aging is cell senescence, a state in which cells remain viable but lose their proliferative capacity, undergo metabolic alterations, and become resistant to apoptosis. This is accompanied by complex cellular changes that enable the development of a senescence-associated secretory phenotype (SASP). Mitochondria, organelles involved in energy provision and activities essential for regulating cell survival and death, are negatively impacted by aging. The age-associated decline in mitochondrial function is also accompanied by the development of chronic low-grade sterile inflammation. The latter shares some features and mediators with the SASP. Indeed, the unloading of damage-associated molecular patterns (DAMPs) at the extracellular level can trigger sterile inflammatory responses and mitochondria can contribute to the generation of DAMPs with pro-inflammatory properties. The extrusion of mitochondrial DNA (mtDNA) via mitochondrial outer membrane permeabilization under an apoptotic stress triggers senescence programs. Additional pathways can contribute to sterile inflammation. For instance, pyroptosis is a caspase-dependent inducer of systemic inflammation, which is also elicited by mtDNA release and contributes to aging. Herein, we overview the molecular mechanisms that may link mitochondrial dyshomeostasis, pyroptosis, sterile inflammation, and senescence and discuss how these contribute to aging and could be exploited as molecular targets for alleviating the cell damage burden and achieving healthy longevity.


Sujet(s)
Survie cellulaire , Vieillissement de la cellule , Mitochondries , Transduction du signal , Humains , Mitochondries/métabolisme , Animaux , ADN mitochondrial/métabolisme , ADN mitochondrial/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Mort cellulaire , Apoptose , Pyroptose , Vieillissement/métabolisme
20.
J Am Heart Assoc ; 13(14): e032904, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38979831

RÉSUMÉ

BACKGROUND: Cardiac aging represents an independent risk factor for aging-associated cardiovascular diseases. Although evidence suggests an association between NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome formation and numerous cardiovascular diseases, its role in cardiac aging remains largely unclear. METHODS AND RESULTS: The longevity of mice with wild-type and NLRP3 knockout (NLRP3-/-) genotypes was assessed, with or without d-galactose treatment. Cardiac function was evaluated using echocardiography, and cardiac histopathology was examined through hematoxylin and eosin and Masson's trichrome staining. Senescence-associated ß-galactosidase (SA-ß-gal) staining was employed to detect cardiac aging. Western blotting was used to assess aging-related proteins (p53, p21) and pyroptosis-related proteins. Additionally, dihydroethidium staining, lactate dehydrogenase release, and interleukin-1ß ELISA assays were performed, along with measurements of total superoxide dismutase and malondialdehyde levels. In vitro, H9c2 cells were exposed to d-galactose for 24 hours in the absence or presence of N-acetyl-l-cysteine (reactive oxygen species inhibitor), BAY-117082 (nuclear factor κ-light-chain enhancer of activated B cells inhibitor), MCC950 (NLRP3 inhibitor), and VX-765 (Caspase-1 inhibitor). Immunofluorescence staining was employed to detect p53, gasdermin D, and apoptosis-associated speck-like protein proteins. Intracellular reactive oxygen species levels were assessed using fluorescence microscopy and flow cytometry. Senescence-associated ß-galactosidase staining and Western blotting were also employed in vitro for the same purpose. The results showed that NLRP3 upregulation was implicated in aging and cardiovascular diseases. Inhibition of NLRP3 extended life span, mitigated the aging phenotype, improved cardiac function and blood pressure, ameliorated lipid metabolism abnormalities, inhibited pyroptosis in cardiomyocytes, and ultimately alleviated cardiac aging. In vitro, the inhibition of reactive oxygen species, nuclear factor κ-light-chain enhancer of activated B cells, NLRP3, or caspase-1 attenuated NLRP3 inflammasome-mediated pyroptosis. CONCLUSIONS: The reactive oxygen species/nuclear factor κ-light-chain enhancer of activated B cells/NLRP3 signaling pathway loop contributes to d-galactose-treated cardiomyocyte senescence and cardiac aging.


Sujet(s)
Galactose , Inflammasomes , Souris knockout , Myocytes cardiaques , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Galactose/toxicité , Galactose/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Inflammasomes/métabolisme , Souris , Vieillissement/métabolisme , Souris de lignée C57BL , Transduction du signal , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Mâle , Espèces réactives de l'oxygène/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire , Modèles animaux de maladie humaine , Rats
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...