Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 808
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.
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
3.
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
4.
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
5.
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
6.
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
7.
Signal Transduct Target Ther ; 9(1): 181, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38992067

RÉSUMÉ

Mitotic catastrophe (MC), which occurs under dysregulated mitosis, represents a fascinating tactic to specifically eradicate tumor cells. Whether pyroptosis can be a death form of MC remains unknown. Proteasome-mediated protein degradation is crucial for M-phase. Bortezomib (BTZ), which inhibits the 20S catalytic particle of proteasome, is approved to treat multiple myeloma and mantle cell lymphoma, but not solid tumors due to primary resistance. To date, whether and how proteasome inhibitor affected the fates of cells in M-phase remains unexplored. Here, we show that BTZ treatment, or silencing of PSMC5, a subunit of 19S regulatory particle of proteasome, causes G2- and M-phase arrest, multi-polar spindle formation, and consequent caspase-3/GSDME-mediated pyroptosis in M-phase (designated as mitotic pyroptosis). Further investigations reveal that inhibitor of WEE1/PKMYT1 (PD0166285), but not inhibitor of ATR, CHK1 or CHK2, abrogates the BTZ-induced G2-phase arrest, thus exacerbates the BTZ-induced mitotic arrest and pyroptosis. Combined BTZ and PD0166285 treatment (named BP-Combo) selectively kills various types of solid tumor cells, and significantly lessens the IC50 of both BTZ and PD0166285 compared to BTZ or PD0166285 monotreatment. Studies using various mouse models show that BP-Combo has much stronger inhibition on tumor growth and metastasis than BTZ or PD0166285 monotreatment, and no obvious toxicity is observed in BP-Combo-treated mice. These findings disclose the effect of proteasome inhibitors in inducing pyroptosis in M-phase, characterize pyroptosis as a new death form of mitotic catastrophe, and identify dual inhibition of proteasome and WEE family kinases as a promising anti-cancer strategy to selectively kill solid tumor cells.


Sujet(s)
Bortézomib , Protéines du cycle cellulaire , Mitose , Proteasome endopeptidase complex , Protein-tyrosine kinases , Pyroptose , Pyroptose/effets des médicaments et des substances chimiques , Humains , Souris , Animaux , Protein-tyrosine kinases/génétique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/métabolisme , Mitose/effets des médicaments et des substances chimiques , Mitose/génétique , Proteasome endopeptidase complex/métabolisme , Proteasome endopeptidase complex/génétique , Bortézomib/pharmacologie , Lignée cellulaire tumorale , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Inhibiteurs du protéasome/pharmacologie , Pyrimidines/pharmacologie , Pyrazoles/pharmacologie , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe , Gasdermines , Pyrimidinones
8.
Int J Biol Sci ; 20(9): 3393-3411, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993566

RÉSUMÉ

Chronic prostatitis is one of the most common urologic diseases that troubles young men, with unclear etiology and ineffective treatment approach. Pyroptosis is a novel model of cell death, and its roles in chronic prostatitis are unknown. In this study, P2X7R, NEK7, and GSDMD-NT expression levels were detected in prostate tissues from benign prostate hyperplasia (BPH) patients and experiment autoimmune prostatitis (EAP) mice. P2X7R agonist, antagonist, NLRP3 inhibitor, and disulfiram were used to explore the roles of the P2X7R-NEK7-NLRP3 axis in prostate epithelial cell pyroptosis and chronic prostatitis development. We found that P2X7R, NEK7, and GSDMD-NT were highly expressed in the prostate epithelial cells of BPH patients with prostatic inflammation and EAP mice. Activation of P2X7R exacerbated prostatic inflammation and increased NLRP3 inflammasome component expressions and T helper 17 (Th17) cell proportion. Moreover, P2X7R-mediated potassium efflux promoted NEK7-NLRP3 interaction, and NLRP3 assembly and activation, which caused GSDMD-NT-mediated prostate epithelial cell pyroptosis to exacerbate EAP development. Disulfiram could effectively improve EAP by inhibiting GSDMD-NT-mediated prostate epithelial cell pyroptosis. In conclusion, the P2X7R-NEK7-NLRP3 axis could promote GSDMD-NT-mediated prostate epithelial cell pyroptosis and chronic prostatitis development, and disulfiram may be an effective drug to treat chronic prostatitis.


Sujet(s)
Cellules épithéliales , Kinases apparentées à NIMA , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéines de liaison aux phosphates , Prostate , Prostatite , Pyroptose , Mâle , Animaux , Prostatite/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Kinases apparentées à NIMA/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Souris , Cellules épithéliales/métabolisme , Humains , Protéines de liaison aux phosphates/métabolisme , Prostate/métabolisme , Souris de lignée C57BL , Récepteurs purinergiques P2X7/métabolisme , Protéines liant le poly-adp-ribose/métabolisme , Maladies auto-immunes/métabolisme , Gasdermines
9.
Cells ; 13(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38994991

RÉSUMÉ

Clostridium perfringens (C. perfringens), a Gram-positive bacterium, produces a variety of toxins and extracellular enzymes that can lead to disease in both humans and animals. Common symptoms include abdominal swelling, diarrhea, and intestinal inflammation. Severe cases can result in complications like intestinal hemorrhage, edema, and even death. The primary toxins contributing to morbidity in C. perfringens-infected intestines are CPA, CPB, CPB2, CPE, and PFO. Amongst these, CPB, CPB2, and CPE are implicated in apoptosis development, while CPA is associated with cell death, increased intracellular ROS levels, and the release of the inflammatory factor IL-18. However, the exact mechanism by which PFO toxins exert their effects in the infected gut is still unidentified. This study demonstrates that a C. perfringens PFO toxin infection disrupts the intestinal epithelial barrier function through in vitro and in vivo models. This study emphasizes the notable influence of PFO toxins on intestinal barrier integrity in the context of C. perfringens infections. It reveals that PFO toxins increase ROS production by causing mitochondrial damage, triggering pyroptosis in IPEC-J2 cells, and consequently resulting in compromised intestinal barrier function. These results offer a scientific foundation for developing preventive and therapeutic approaches against C. perfringens infections.


Sujet(s)
Toxines bactériennes , Clostridium perfringens , Cellules épithéliales , Hémolysines , Muqueuse intestinale , Pyroptose , Espèces réactives de l'oxygène , Clostridium perfringens/pathogénicité , Toxines bactériennes/toxicité , Toxines bactériennes/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Animaux , Hémolysines/métabolisme , Hémolysines/toxicité , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/microbiologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire , Souris , Humains , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques
10.
PLoS One ; 19(7): e0298353, 2024.
Article de Anglais | MEDLINE | ID: mdl-38995910

RÉSUMÉ

CONTEXT: Nephrotic syndrome(NS) has emerged as a worldwide public health problem. Renal fibrosis is the most common pathological change from NS to end-stage renal failure, seriously affecting the prognosis of renal disease. Although tremendous efforts have been made to treat NS, specific drug therapies to delay the progression of NS toward end-stage renal failure are limited. Epimedium is generally used to treat kidney disease in traditional Chinese medicine. Icariin is a principal active component of Epimedium. METHODS: We used Sprague Dawley rats to establish NS models by injecting doxorubicin through the tail vein. Then icariin and prednisone were intragastric administration. Renal function was examined by an automatic biochemical analyzer. Pathology of the kidney was detected by Hematoxylin-Eosin and Masson staining respectively. Furthermore, RT-PCR, Enzyme-Linked Immunosorbent Assay, Immunohistochemistry, Western Blot and Terminal-deoxynucleotidyl Transferase Mediated Nick End Labeling staining were employed to detect the proteins related to pyroptosis and EMT. HK-2 cells exposed to doxorubicin were treated with icariin, and cell viability was assessed using the MTT. EMT was assessed using Enzyme-Linked Immunosorbent Assay and Western Blot. RESULTS: The study showed that icariin significantly improved renal function and renal fibrosis in rats. In addition, icariin effectively decreased NOD-like receptor thermal protein domain associated protein 3,Caspase-1, Gasdermin D, Ly6C, and interleukin (IL)-1ß. Notably, treatment with icariin also inhibited the levels of TGF-ß, α-SMA and E-cadherin. DISCUSSION AND CONCLUSIONS: It is confirmed that icariin can improve renal function and alleviate renal fibrosis by inhibiting pyroptosis and the mechanism may be related to epithelial-to-mesenchymal transition. Icariin treatment might be recommended as a new approach for NS.


Sujet(s)
Doxorubicine , Transition épithélio-mésenchymateuse , Flavonoïdes , Syndrome néphrotique , Pyroptose , Rat Sprague-Dawley , Animaux , Flavonoïdes/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Pyroptose/effets des médicaments et des substances chimiques , Rats , Syndrome néphrotique/traitement médicamenteux , Syndrome néphrotique/anatomopathologie , Syndrome néphrotique/métabolisme , Mâle , Doxorubicine/pharmacologie , Humains , Fibrose/traitement médicamenteux , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Lignée cellulaire , Modèles animaux de maladie humaine
11.
Mol Biol Rep ; 51(1): 802, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39001949

RÉSUMÉ

BACKGROUND: Alzheimer's disease is a neurological disease characterized by the build-up of amyloid beta peptide (Aß) and lipopolysaccharide (LPS), which causes synapse dysfunction, cell death, and neuro-inflammation. A maladaptive unfolded protein response (UPR), excessive autophagy, and pyroptosis aggravate the disease. Melatonin (MEL) and hydroxybutyrate (BHB) have both shown promise in terms of decreasing Aß pathology. The goal of this study was to see how BHB and MEL affected the UPR, autophagy, and pyroptosis pathways in Aß1-42 and LPS-induced SH-SY5Y cells. MATERIALS AND METHODS: Human neuroblastoma SH-SY5Y cells were treated with BHB, MEL, or a combination of the two after being exposed to A ß1-42 and LPS. Cell viability was determined using the MTT test, and gene expression levels of UPR (ATF6, PERK, and CHOP), autophagy (Beclin-1, LC3II, P62, and Atg5), and pyroptosis-related markers (NLRP3, TXNIP, IL-1ß, and NFκB1) were determined using quantitative Real-Time PCR (qRT-PCR). For statistical analysis, one-way ANOVA was employed, followed by Tukey's post hoc test. RESULTS: BHB and MEL significantly increased SH-SY5Y cell viability in the presence of A ß1-42 and LPS. Both compounds inhibited the expression of maladaptive UPR and autophagy-related genes, as well as inflammatory and pyroptotic markers caused by Aß1-42 and LPS-induced SH-SY5Y cells. CONCLUSION: BHB and MEL rescue neurons in A ß1-42 and LPS-induced SH-SY5Y cells by reducing maladaptive UPR, excessive autophagy, and pyroptosis. More research is needed to fully comprehend the processes behind their beneficial effects and to discover their practical applications in the treatment of neurodegenerative disorders.


Sujet(s)
Acide 3-hydroxy-butyrique , Peptides bêta-amyloïdes , Autophagie , Lipopolysaccharides , Mélatonine , Fragments peptidiques , Pyroptose , Réponse aux protéines mal repliées , Humains , Mélatonine/pharmacologie , Peptides bêta-amyloïdes/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Pyroptose/effets des médicaments et des substances chimiques , Lipopolysaccharides/pharmacologie , Lignée cellulaire tumorale , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Acide 3-hydroxy-butyrique/pharmacologie , Fragments peptidiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/traitement médicamenteux , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie
12.
J Nanobiotechnology ; 22(1): 393, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965602

RÉSUMÉ

BACKGROUND: The therapeutic strategies for acute ischemic stroke were faced with substantial constraints, emphasizing the necessity to safeguard neuronal cells during cerebral ischemia to reduce neurological impairments and enhance recovery outcomes. Despite its potential as a neuroprotective agent in stroke treatment, Chikusetsu saponin IVa encounters numerous challenges in clinical application. RESULT: Brain-targeted liposomes modified with THRre peptides showed substantial uptake by bEnd. 3 and PC-12 cells and demonstrated the ability to cross an in vitro blood-brain barrier model, subsequently accumulating in PC-12 cells. In vivo, they could significantly accumulate in rat brain. Treatment with C-IVa-LPs-THRre notably reduced the expression of proteins in the P2RX7/NLRP3/Caspase-1 pathway and inflammatory factors. This was evidenced by decreased cerebral infarct size and improved neurological function in MCAO rats. CONCLUSION: The findings indicate that C-IVa-LPs-THRre could serve as a promising strategy for targeting cerebral ischemia. This approach enhances drug concentration in the brain, mitigates pyroptosis, and improves the neuroinflammatory response associated with stroke.


Sujet(s)
Barrière hémato-encéphalique , Accident vasculaire cérébral ischémique , Liposomes , Neuroprotecteurs , Pyroptose , Rat Sprague-Dawley , Saponines , Animaux , Saponines/pharmacologie , Saponines/composition chimique , Pyroptose/effets des médicaments et des substances chimiques , Rats , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Liposomes/composition chimique , Mâle , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Cellules PC12 , Acide oléanolique/pharmacologie , Acide oléanolique/composition chimique , Acide oléanolique/analogues et dérivés , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Peptides/composition chimique , Peptides/pharmacologie , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme
13.
Sci Rep ; 14(1): 16270, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009650

RÉSUMÉ

Steroid-induced osteonecrosis of the femoral head (SONFH) is the predominant cause of non-traumatic osteonecrosis of the femoral head (ONFH). Impaired blood supply and reduced osteogenic activity of the femoral head are the key pathogenic mechanisms of SONFH. Fibroblast growth factor 23 (FGF23) levels are not only a biomarker for early vascular lesions caused by abnormal mineral metabolism, but can also act directly on the peripheral vascular system, leading to vascular pathology. The aim of this study was to observe the role of FGF23 on bone microarchitecture and vascular endothelium, and to investigate activation of pyroptosis in SONFH. Lipopolysaccharide (LPS) combined with methylprednisolone (MPS) was applied for SONFH mouse models, and adenovirus was used to increase or decrease the level of FGF23. Micro-CT and histopathological staining were used to observe the structure of the femoral head, and immunohistochemical staining was used to observe the vascular density. The cells were further cultured in vitro and placed in a hypoxic environment for 12 h to simulate the microenvironment of vascular injury during SONFH. The effect of FGF23 on osteogenic differentiation was evaluated using alkaline phosphatase staining, alizarin red S staining and expression of bone formation-related proteins. Matrigel tube formation assay in vitro and immunofluorescence were used to detect the ability of FGF23 to affect endothelial cell angiogenesis. Steroids activated the pyroptosis signaling pathway, promoted the secretion of inflammatory factors in SONFH models, led to vascular endothelial dysfunction and damaged the femoral head structure. In addition, FGF23 inhibited the HUVECs angiogenesis and BMSCs osteogenic differentiation. FGF23 silencing attenuated steroid-induced osteonecrosis of the femoral head by inhibiting the pyroptosis signaling pathway, and promoting osteogenic differentiation of BMSCs and angiogenesis of HUVECs in vitro.


Sujet(s)
Nécrose de la tête fémorale , Facteur-23 de croissance des fibroblastes , Facteurs de croissance fibroblastique , Ostéogenèse , Pyroptose , Pyroptose/effets des médicaments et des substances chimiques , Facteur-23 de croissance des fibroblastes/métabolisme , Animaux , Nécrose de la tête fémorale/induit chimiquement , Nécrose de la tête fémorale/métabolisme , Nécrose de la tête fémorale/anatomopathologie , Souris , Facteurs de croissance fibroblastique/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Humains , Tête du fémur/anatomopathologie , Tête du fémur/métabolisme , Modèles animaux de maladie humaine , Méthylprednisolone/pharmacologie , Mâle , Lipopolysaccharides/toxicité , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Différenciation cellulaire , Stéroïdes/pharmacologie
14.
Molecules ; 29(12)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38930963

RÉSUMÉ

Ulcerative colitis (UC) is difficult to cure and easy to relapse, leading to poor quality of life for patients. Oxymatrine (OMT) is one of the main alkaloids of Sophora flavescens Aiton, which has many effects, such as anti-inflammation, anti-oxidative stress, and immunosuppression. This study aimed to investigate whether OMT could attenuate ulcerative colitis by inhibiting the NOD-like receptor family pyrin domain containing three (NLRP3) inflammasome-mediated pyroptosis. In this study, the UC rat models were established by 2,4,6-Trinitrobenzenesulfonic acid (TNBS) in vivo, while RAW264.7 cells and peritoneal macrophages were stimulated with Lipopolysaccharides/Adenosine Triphosphate (LPS/ATP) in vitro to simulate pyroptosis models, and Western blotting (WB) and other detection techniques were applied to analyze proteins involved in the NLRP3 inflammasome pathway. Our results showed that OMT alleviated colitis ulcers and pathological damage in the TNBS-induced UC rats and exhibited an inhibitory effect on pyroptosis at the early stage of UC. In the model group, the pyroptosis reached the peak at 24 h after modeling with the contents of active-cysteine-aspartic proteases-1 (caspase-1), Gasdermin D (GSDMD)-N, and cleaved-interleukin-1 beta (IL-1ß) to the highest expression level. Meanwhile, we found that OMT (80 mg kg-1) remarkably decreased the expression levels of NLRP3, active-caspase-1, and cleaved-IL-1ß at 24 h in the lesion tissue from UC rats. Further experiments on cells demonstrated that OMT at concentrations of 100 and 250 µM significantly inhibited cell death caused by NLRP3 inflammasome activation (p < 0.05), downregulated caspase-1, GSDMD, and decreased the levels of active-caspase-1, GSDMD-N, cleaved-IL-1ß in RAW326.7 cells, and peritoneal macrophages. In summary, these results indicated that OMT could attenuate ulcerative colitis through inhibiting pyroptosis mediated by the NLRP3 inflammasome. The inhibition of the NLRP3 inflammasome may be a potential strategy for UC.


Sujet(s)
Alcaloïdes , Rectocolite hémorragique , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Quinolizines , Animaux , Quinolizines/pharmacologie , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Alcaloïdes/pharmacologie , Pyroptose/effets des médicaments et des substances chimiques , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Souris , Rats , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Cellules RAW 264.7 , Mâle , Modèles animaux de maladie humaine , Rat Sprague-Dawley , Acide 2,4,6-trinitro-benzènesulfonique , Lipopolysaccharides ,
15.
Phytomedicine ; 130: 155756, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38833791

RÉSUMÉ

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a persistent liver condition that affects both human health and animal productive efficiency on a global scale. A number of naturally occurring compounds activate nuclear factor erythroid 2-related factor 2 (Nrf2) as a transcription factor with important protective effects against many liver diseases, including NAFLD. Raffinose (Ra), an oligosaccharide extracted from several plants, exhibits diverse biological functions. However, the uncertainty lies in determining whether the activation of Nrf2 by Ra can provide a preventive effect on liver lipotoxicity. PURPOSE: The aim of this study was to shed light on the molecular pathways by which Ra possesses its protective benefits against NAFLD. METHODS: Experimental protocols were established using WT and Nrf2-null (Nrf2-/-) mice. Liver samples from each group were collected for Western blot, RT-qPCR, H & E, Sirius red and Oil red O staining. Additionally, serums were processed for ELISA. ALM12 cells were gathered for Western blot and immunofluorescence. Moreover, to elucidate the molecular mechanism of Ra, molecular docking was performed. RESULTS: Our results indicated that Ra remarkably alleviated liver lipotoxic in vivo and in vitro. Ra treatment effectively corrected hepatic steatosis, the release of AST, ALT, TG, and TC, as well as the depletion of HDL and LDL. Meanwhile, Ra efficiently prevented inflammation by inhibiting the TLR4-MyD88-NF-κB pathway and pyroptosis. Additionally, these findings implied that Ra reduced the production of fibrosis-related proteins, which enhanced collagen deposition. Molecular docking revealed that Ra possessed the ability to bind specific regions of Nrf2, resulting in the enhancement of Nrf2 activation and nuclear translocation. Ra treatment restored serum redox factors and antioxidant enzymes to normal levels; however, these alterations were clearly reversed in Nrf2-/- mice. CONCLUSION: This study reveals novel information on Ra's protective benefits against liver injury caused by abnormal lipid metabolism; these effects are mostly mediated by Nrf2 activation, suggesting a potential new medicine or treatment strategy for NAFLD.


Sujet(s)
Facteur-2 apparenté à NF-E2 , Stéatose hépatique non alcoolique , Pyroptose , Animaux , Stéatose hépatique non alcoolique/traitement médicamenteux , Facteur-2 apparenté à NF-E2/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Souris , Récepteur de type Toll-4/métabolisme , Mâle , Métabolisme lipidique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Inflammation/traitement médicamenteux , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Simulation de docking moléculaire , Antioxydants/pharmacologie , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/métabolisme , Souris knockout , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de différenciation myéloïde-88/métabolisme , Facteur de transcription NF-kappa B/métabolisme
16.
PLoS One ; 19(6): e0305409, 2024.
Article de Anglais | MEDLINE | ID: mdl-38875245

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Pulmonary fibrosis caused by lung injury is accompanied by varying degrees of inflammation, and diazepam can reduce the levels of inflammatory factors. Therefore, the purpose of this study was to determine whether diazepam can inhibit inflammation and ameliorate pulmonary fibrosis by regulating the let-7a-5p/myeloid differentiation factor 88 (MYD88) axis. METHODS: Lipopolysaccharide (LPS) was used to induce cell pyroptosis in an animal model of pulmonary fibrosis. After treatment with diazepam, changes in cell proliferation and apoptosis were observed, and the occurrence of inflammation and pulmonary fibrosis in the mice was detected. RESULTS: The results showed that LPS can successfully induce cell pyroptosis and inflammatory responses and cause lung fibrosis in mice. Diazepam inhibits the expression of pyroptosis-related factors and inflammatory factors; moreover, it attenuates the occurrence of pulmonary fibrosis in mice. Mechanistically, diazepam can upregulate the expression of let-7a-5p, inhibit the expression of MYD88, and reduce inflammation and inhibit pulmonary fibrosis by regulating the let-7a-5p/MYD88 axis. CONCLUSION: Our findings indicated that diazepam can inhibit LPS-induced pyroptosis and inflammatory responses and alleviate pulmonary fibrosis in mice by regulating the let-7a-5p/MYD88 axis.


Sujet(s)
Diazépam , Inflammation , Lipopolysaccharides , microARN , Facteur de différenciation myéloïde-88 , Fibrose pulmonaire , Pyroptose , Animaux , Pyroptose/effets des médicaments et des substances chimiques , Souris , Diazépam/pharmacologie , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , microARN/génétique , microARN/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques
17.
Int J Nanomedicine ; 19: 4957-4976, 2024.
Article de Anglais | MEDLINE | ID: mdl-38828198

RÉSUMÉ

Background: The "gut-islets axis" is an important endocrine signaling axis that regulates islets function by modulating the gut microbiota and endocrine metabolism within the gut. However, the specific mechanisms and roles of the intestine in islets regulation remain unclear. Recent studies investigated that exosomes derived from gut microbiota can transport signals to remotely regulate islets ß-cell function, suggesting the possibility of novel signaling pathways mediated by gut exosomes in the regulation of the "gut-islet axis.". Methods: The exosomes were isolated from the intestinal enteroendocrine cell-line STC-1cells culture supernatants treated with palmitate acid (PA) or BSA. Metabolic stress models were established by separately subjecting MIN6 cells to PA stimulation and feeding mice with a high-fat diet. Intervention with exosomes in vitro and in vivo to assess the biological effects of exosomes on islets ß cells under metabolic stress. The Mas receptor antagonist A779 and ACE2ko mice were used to evaluate the role of exosomal ACE2. Results: We found ACE2, a molecule that plays a crucial role in the regulation of islets function, is abundantly expressed in exosomes derived from STC-1 under physiological normal condition (NCEO). These exosomes cannot only be taken up by ß-cells in vitro but also selectively transported to the islets in vivo. Following intervention with NCEXO, both Min6 cells in a lipotoxic environment and mice on a high-fat diet exhibited significant improvements in islets ß-cell function and ß-cell mass. Further investigations demonstrated that these protective effects are attributed to exosomal ACE2, as ACE2 inhibits NLRP3 inflammasome activation and reduces ß-cell pyroptosis. Conclusion: ACE2-enriched exosomes from the gut can selectively target islets, subsequently inhibiting NLRP3 inflammasome activation and ß cell pyroptosis, thereby restoring islets ß cell function under metabolic stress. This study provides novel insights into therapeutic strategies for the prevention and treatment of obesity and diabetes.


Sujet(s)
Angiotensin-converting enzyme 2 , Exosomes , Inflammasomes , Cellules à insuline , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Animaux , Exosomes/métabolisme , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Souris , Pyroptose/effets des médicaments et des substances chimiques , Pyroptose/physiologie , Angiotensin-converting enzyme 2/métabolisme , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Lignée cellulaire , Intestin grêle/effets des médicaments et des substances chimiques , Mâle , Alimentation riche en graisse , Souris knockout , Cellules entéroendocrines/effets des médicaments et des substances chimiques , Cellules entéroendocrines/métabolisme
18.
Pestic Biochem Physiol ; 202: 105930, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38879323

RÉSUMÉ

Due to the widespread use of metolachlor (MET), the accumulation of MET and its metabolites in the environment has brought serious health problems to aquatic organisms. At present, the toxicity of MET on the physiological metabolism of aquatic animals mainly focused on the role of enzymes. There is still a lack of research on the molecular mechanisms of MET hepatotoxicity, especially on antagonizing MET toxicity. Therefore, this study focuses on grass carp hepatocytes (L8824 cells) closely related to toxin accumulation. By establishing a MET exposed L8824 cells model, it is determined that MET exposure induces pyrolytic inflammation of L8824 cells. Subsequent mechanistic studies found that MET exposure induces pyroptosis in L8824 cells through mitochondrial dysfunction, and siCaspase-1 inhibits the MET induced ROS production, suggesting a regulation of ROS-NLRP3- Caspase-1 pyroptotic inflammation cycling center in MET induced injury to L8824 cells. Molecular docking revealed a strong binding energy between melatonin (MT) and Caspase-1. Finally, a model of L8824 cells with MT intervention in MET exposure was established. MT can antagonize the pyroptosis induced by MET exposure in L8824 cells by targeting Caspase-1, thereby restoring mitochondrial function and inhibiting the ROS-pyroptosis cycle. This study discovered targets and mechanisms of MT regulating pyroptosis in MET exposed-L8824 cells, and the results are helpful to provide new targets for the design of MET antidotes.


Sujet(s)
Acétamides , Carpes (poisson) , Hépatocytes , Mélatonine , Simulation de docking moléculaire , Animaux , Carpes (poisson)/métabolisme , Mélatonine/pharmacologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Acétamides/toxicité , Acétamides/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire , Pyroptose/effets des médicaments et des substances chimiques , Caspase-1/métabolisme , Herbicides/toxicité , Simulation numérique , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme
19.
Mol Med Rep ; 30(2)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38873985

RÉSUMÉ

Macrophage pyroptosis mediates vascular inflammation and atherosclerosis (AS). Hydrogen sulfide (H2S) exerts a protective role in preventing inflammation and AS. However, its molecular mechanisms of regulating the pyroptosis signaling pathway and inhibiting macrophage pyroptosis remain unexplored. The present study aimed to determine whether H2S mitigates macrophage pyroptosis by downregulating the pyroptosis signaling pathway and S­sulfhydrating caspase­1 under the stimulation of oxidized low­density lipoprotein (ox­LDL), a pro­atherosclerotic factor. Macrophages derived from THP­1 monocytes were pre­treated using exogenous H2S donors sodium hydrosulfide (NaHS) and D,L­propargylglycine (PAG), a pharmacological inhibitor of endogenous H2S­producing enzymes, alone or in combination. Subsequently, cells were stimulated with ox­LDL or the desulfhydration reagent dithiothreitol (DTT) in the presence or absence of NaHS and/or PAG. Following treatment, the levels of H2S in THP­1 derived macrophages were measured by a methylene blue colorimetric assay. The pyroptotic phenotype of THP­1 cells was observed and evaluated by light microscopy, Hoechst 33342/propidium iodide fluorescent staining and lactate dehydrogenase (LDH) release assay. Caspase­1 activity in THP­1 cells was assayed by caspase­1 activity assay kit. Immunofluorescence staining was used to assess the accumulation of active caspase­1. Western blotting and ELISA were performed to determine the expression of pyroptosis­specific markers (NLRP3, pro­caspase­1, caspase­1, GSDMD and GSDMD­N) in cells and the secretion of pyroptosis­related cytokines [interleukin (IL)­1ß and IL­18] in the cell­free media, respectively. The S­sulfhydration of pro­caspase­1 in cells was assessed using a biotin switch assay. ox­LDL significantly induced macrophage pyroptosis by activating the pyroptosis signaling pathway. Inhibition of endogenous H2S synthesis by PAG augmented the pro­pyroptotic effects of ox­LDL. Conversely, exogenous H2S (NaHS) ameliorated ox­LDL­and ox­LDL + PAG­induced macrophage pyroptosis by suppressing the activation of the pyroptosis signaling pathway. Mechanistically, ox­LDL and the DTT increased caspase­1 activity and downstream events (IL­1ß and IL­18 secretion) of the caspase­1­dependent pyroptosis pathway by reducing S­sulfhydration of pro­caspase­1. Conversely, NaHS increased S­sulfhydration of pro­caspase­1, reducing caspase­1 activity and caspase­1­dependent macrophage pyroptosis. The present study demonstrated the molecular mechanism by which H2S ameliorates macrophage pyroptosis by suppressing the pyroptosis signaling pathway and S­sulfhydration of pro­caspase­1, thereby suppressing the generation of active caspase-1 and activity of caspase-1.


Sujet(s)
Caspase-1 , Sulfure d'hydrogène , Lipoprotéines LDL , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéines de liaison aux phosphates , Pyroptose , Sulfure d'hydrogène/pharmacologie , Sulfure d'hydrogène/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Humains , Caspase-1/métabolisme , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Lipoprotéines LDL/métabolisme , Lipoprotéines LDL/pharmacologie , Protéines de liaison aux phosphates/métabolisme , Cellules THP-1 , Protéines et peptides de signalisation intracellulaire/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Gasdermines , Alcynes , Glycine/analogues et dérivés , Sulfures
20.
Int J Nanomedicine ; 19: 5545-5579, 2024.
Article de Anglais | MEDLINE | ID: mdl-38882539

RÉSUMÉ

Pyroptosis, a pro-inflammatory and lytic programmed cell death pathway, possesses great potential for antitumor immunotherapy. By releasing cellular contents and a large number of pro-inflammatory factors, tumor cell pyroptosis can promote dendritic cell maturation, increase the intratumoral infiltration of cytotoxic T cells and natural killer cells, and reduce the number of immunosuppressive cells within the tumor. However, the efficient induction of pyroptosis and prevention of damage to normal tissues or cells is an urgent concern to be addressed. Recently, a wide variety of nanoplatforms have been designed to precisely trigger pyroptosis and activate the antitumor immune responses. This review provides an update on the progress in nanotechnology for enhancing pyroptosis-based tumor immunotherapy. Nanomaterials have shown great advantages in triggering pyroptosis by delivering pyroptosis initiators to tumors, increasing oxidative stress in tumor cells, and inducing intracellular osmotic pressure changes or ion imbalances. In addition, the challenges and future perspectives in this field are proposed to advance the clinical translation of pyroptosis-inducing nanomedicines.


Sujet(s)
Immunothérapie , Nanostructures , Tumeurs , Pyroptose , Pyroptose/effets des médicaments et des substances chimiques , Humains , Immunothérapie/méthodes , Tumeurs/thérapie , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Nanostructures/composition chimique , Animaux , Nanomédecine , Stress oxydatif/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...