Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 973
Filter
1.
J Environ Sci (China) ; 148: 13-26, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39095152

ABSTRACT

Bisphenol A (BPA) is an industrial pollutant that can cause immune impairment. Selenium acts as an antioxidant, as selenium deficiency often accompanies oxidative stress, resulting in organ damage. This study is the first to demonstrate that BPA and/or selenium deficiency induce pyroptosis and ferroptosis-mediated thymic injury in chicken and chicken lymphoma cell (MDCC-MSB-1) via oxidative stress-induced endoplasmic reticulum (ER) stress. We established a broiler chicken model of BPA and/or selenium deficiency exposure and collected thymus samples as research subjects after 42 days. The results demonstrated that BPA or selenium deficiency led to a decrease in antioxidant enzyme activities (T-AOC, CAT, and GSH-Px), accumulation of peroxides (H2O2 and MDA), significant upregulation of ER stress-related markers (GRP78, IER 1, PERK, EIF-2α, ATF4, and CHOP), a significant increase in iron ion levels, significant upregulation of pyroptosis-related gene (NLRP3, ASC, Caspase1, GSDMD, IL-18 and IL-1ß), significantly increase ferroptosis-related genes (TFRC, COX2) and downregulate GPX4, HO-1, FTH, NADPH. In vitro experiments conducted in MDCC-MSB-1 cells confirmed the results, demonstrating that the addition of antioxidant (NAC), ER stress inhibitor (TUDCA) and pyroptosis inhibitor (Vx765) alleviated oxidative stress, endoplasmic reticulum stress, pyroptosis, and ferroptosis. Overall, this study concludes that the combined effects of oxidative stress and ER stress mediate pyroptosis and ferroptosis in chicken thymus induced by BPA exposure and selenium deficiency.


Subject(s)
Benzhydryl Compounds , Chickens , Endoplasmic Reticulum Stress , Ferroptosis , Phenols , Pyroptosis , Reactive Oxygen Species , Selenium , Animals , Benzhydryl Compounds/toxicity , Ferroptosis/drug effects , Pyroptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Selenium/deficiency , Phenols/toxicity , Reactive Oxygen Species/metabolism , Thymus Gland/drug effects , Oxidative Stress/drug effects
2.
World J Gastroenterol ; 30(34): 3850-3855, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39350786

ABSTRACT

In this editorial, we comment on the article by Zhou et al. The study reveals the connection between ferroptosis and pyroptosis and the effect of silent information regulator sirtuin 1 (SIRT1) activation in acute liver failure (ALF). ALF is characterized by a sudden and severe liver injury resulting in significant hepatocyte damage, often posing a high risk of mortality. The predominant form of hepatic cell death in ALF involves apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Glutathione peroxidase 4 (GPX4) inhibition sensitizes the cell to ferroptosis and triggers cell death, while Gasdermin D (GSDMD) is a mediator of pyroptosis. The study showed that ferroptosis and pyroptosis in ALF are regulated by blocking the p53/GPX4/GSDMD pathway, bridging the gap between the two processes. The inhibition of p53 elevates the levels of GPX4, reducing the levels of inflammatory and liver injury markers, ferroptotic events, and GSDMD-N protein levels. Reduced p53 expression and increased GPX4 on deletion of GSDMD indicated ferroptosis and pyroptosis interaction. SIRT1 is a NAD-dependent deacetylase, and its activation attenuates liver injury and inflammation, accompanied by reduced ferroptosis and pyroptosis-related proteins in ALF. SIRT1 activation also inhibits the p53/GPX4/GSDMD axis by inducing p53 acetylation, attenuating LPS/D-GalN-induced ALF.


Subject(s)
Ferroptosis , Intracellular Signaling Peptides and Proteins , Liver Failure, Acute , Phosphate-Binding Proteins , Phospholipid Hydroperoxide Glutathione Peroxidase , Sirtuin 1 , Tumor Suppressor Protein p53 , Sirtuin 1/metabolism , Sirtuin 1/genetics , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Tumor Suppressor Protein p53/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Ferroptosis/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Animals , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Signal Transduction , Pyroptosis/drug effects , Hepatocytes/metabolism , Liver/pathology , Liver/metabolism , Mice , Gasdermins
3.
World J Gastroenterol ; 30(33): 3791-3798, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39351426

ABSTRACT

In this editorial, we comment on the article published in the recent issue of the World Journal of Gastroenterology. Acute liver failure (ALF) is a fatal disease that causes uncontrolled massive hepatocyte death and rapid loss of liver function. Ferroptosis and pyroptosis, cell death forms that can be initiated or blocked concurrently, can play significant roles in developing inflammation and various malignancies. However, their roles in ALF remain unclear. The article discovered the positive feedback between ferroptosis and pyroptosis in the progression of ALF, and revealed that the silent information regulator sirtuin 1 (SIRT1) inhibits both pathways through p53, dramatically reducing inflammation and protecting hepatocytes. This suggests the potential use of SIRT1 and its downstream molecules as therapeutics for ALF. Thus, we will discuss the role of ferroptosis and pyroptosis in ALF and the crosstalk between these cell death mechanisms. Additionally, we address potential treatments that could alleviate ALF by simultaneously inhibiting both cell death pathways, as well as examples of SIRT1 activators being used as disease treatment strategies, providing new insights into the therapy of ALF.


Subject(s)
Ferroptosis , Hepatocytes , Liver Failure, Acute , Pyroptosis , Sirtuin 1 , Humans , Pyroptosis/drug effects , Ferroptosis/drug effects , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Sirtuin 1/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/drug effects , Signal Transduction/drug effects , Animals , Liver/pathology , Liver/metabolism , Liver/drug effects , Molecular Targeted Therapy/methods , Tumor Suppressor Protein p53/metabolism
4.
Cell Death Dis ; 15(10): 725, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358349

ABSTRACT

Pancreatic cancer is one of the leading causes of cancer-associated mortality, with a poor treatment approach. Previous study has shown that inducing pyroptosis in pancreatic ductal adenocarcinoma (PDAC) slows the growth of PDACs, implying that pyroptosis inducers are potentially effective for PDAC therapy. Here, we found that Dronedarone hydrochloride (DH), an antiarrhythmic drug, induces pyroptosis in pancreatic cancer cells and inhibits PDAC development in mice. In PANC-1 cells, DH caused cell death in a dosage- and time-dependent manner, with only pyroptosis inhibitors and GSDMD silencing rescuing the cell death, indicating that DH triggered GSDMD-dependent pyroptosis. Further work revealed that DH increased mitochondrial stresses and caused mitochondrial DNA (mtDNA) leakage, activating the cytosolic STING-cGAS and pyroptosis pathways. Finally, we assessed the anti-cancer effects of DH in a pancreatic cancer mouse model and found that DH treatment suppressed pancreatic tumor development in vivo. Collectively, our investigation demonstrates that DH triggers pyroptosis in PDAC and proposes its potential effects on anti-PDAC growth.


Subject(s)
DNA, Mitochondrial , Dronedarone , Pancreatic Neoplasms , Pyroptosis , Pyroptosis/drug effects , Animals , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Humans , Dronedarone/pharmacology , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Mice , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/genetics , Mice, Nude
5.
J Immunother Cancer ; 12(10)2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39366751

ABSTRACT

BACKGROUND: In addition to their established action of synthetic lethality in tumor cells, poly(ADP-ribose) polymerase inhibitors (PARPis) also orchestrate tumor immune microenvironment (TIME) that contributes to suppressing tumor growth. However, it remains not fully understood whether and how PARPis trigger tumor-targeting immune responses. METHODS: To decode the immune responses reshaped by PARPis, we conducted T-cell receptor (TCR) sequencing and immunohistochemical (IHC) analyses of paired clinical specimens before and after niraparib monotherapy obtained from a prospective study, as well as ID8 mouse ovarian tumors. To validate the induction of immunogenic cell death (ICD) by PARPis, we performed immunofluorescence/IHC staining with homologous recombination deficiency tumor cells and patient-derived xenograft tumor tissues, respectively. To substantiate that PARPis elicited tumor cell pyroptosis, we undertook comprehensive assessments of the cellular morphological features, cleavage of gasdermin (GSDM) proteins, and activation of TNF-caspase signaling pathways through genetic downregulation/depletion and selective inhibition. We also evaluated the critical role of pyroptosis in tumor suppression and immune activation following niraparib treatment using a syngeneic mouse model with implanting CRISPR/Cas9 edited Gsdme-/ - ID8 tumor cells into C57BL/6 mice. RESULTS: Our findings revealed that PARPis augmented the proportion of neoantigen-recognized TCR clones and TCR clonal expansion, and induced an inflamed TIME characterized by increased infiltration of both innate and adaptive immune cells. This PARPis-strengthened immune response was associated with the induction of ICD, specifically identified as pyroptosis, which possessed distinctive morphological features and GSDMD/E cleavage. It was validated that the cleavage of GSDMD/E was due to elevated caspase 8 activity downstream of the TNFR1, rather than FAS and TRAIL-R. On PARP inhibition, the NF-κB signaling pathway was activated, leading to increased secretion of TNF-α and subsequent initiation of the TNFR1-caspase 8 cascade. Impeding pyroptosis through the depletion of Gsdme significantly compromised the tumor-suppressing effects of PARP inhibition and undermined the anti-immune response in the syngeneic ID8 mouse model. CONCLUSIONS: PARPis induce a specific type of ICD called pyroptosis via TNF-caspase 8-GSDMD/E axis, resulting in an inflamed TIME and augmentation of tumor-targeting immune responses. These findings deepen our understanding of PARPis activities and point toward a promising avenue for synergizing PARPis with immunotherapeutic interventions. TRIAL REGISTRATION NUMBER: NCT04507841.


Subject(s)
Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Pyroptosis , Animals , Female , Humans , Mice , Cell Line, Tumor , Gasdermins , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphate-Binding Proteins/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Pyroptosis/drug effects , Signal Transduction , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism
6.
Front Biosci (Landmark Ed) ; 29(9): 334, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39344337

ABSTRACT

BACKGROUND: Ventilator-induced lung injury (VILI) is a consequence of inflammation and increased alveolar-capillary membrane permeability due to alveolar hyperdistention or elevated intrapulmonary pressure, but the precise mechanisms remain unclear. The aim of the study was to analyze the mechanism by which angiotensin converting enzyme 2 (ACE2) alleviates endoplasmic reticulum stress (ERS) and protects alveolar cells from pyroptosis in VILI by regulating angiotensin (Ang)1-7/Mas. METHODS: VILI was induced in mice by mechanical ventilation by regulating the tidal volume. The alveolar cell line, A549, mimics VILI in vitro by cyclic stretch (CS). Ang (1-7) (100 nmol/L) was added to the medium. ERS was induced in cells by stimulating with tunicamycin (TM, 2 µg/mL). ERS was inhibited by tracheal instillation of 4-phenylbutyric acid (4-PBA) (1 mg/kg). ACE2's enzymatic function was activated or inhibited by subcutaneous injection of resorcinolnaphthalein (RES, 20 µg/kg) or MLN-4760 (20 µg/kg). pGLV-EF1a-GFP-ACE2 was instilled into the trachea to increase the protein expression of ACE2. The Ang (1-7) receptor, Mas, was antagonized by injecting A779 subcutaneously (80 µg/kg). RESULTS: ACE2 protein levels decreased after modeling. Ang (1-7) level was decreased and Ang II was accumulated. ERS was significantly induced in VILI mice, and pyroptosis was observed in cells. When ERS was inhibited, pyroptosis under the VILI condition was significantly inhibited. Ang (1-7) alleviated ERS and pyroptosis under CS. When ERS was continuously activated, the function of Ang (1-7) in inhibiting pyroptosis was blocked. Resorcinolnaphthalein (RES) effectively promoted Ang II conversion, alleviated the Ang (1-7) level in VILI, ameliorated lung injury, and inhibited ERS and cell pyroptosis. Inhibiting ACE2's function in VILI hindered the production of Ang (1-7), promoted the accumulation of Ang II, and exacerbated ERS and pyroptosis, along with lung injury. The Mas antagonist significantly blocked the inhibitory effects of ACE2 on ERS and pyroptosis in VILI. CONCLUSIONS: Reduced ACE2 expression in VILI is involved in ERS and pyroptosis-related injury. ACE2 can alleviate ERS in alveolar cells by catalyzing the production of Ang (1-7), thus inhibiting pyroptosis in VILI.


Subject(s)
Angiotensin I , Angiotensin-Converting Enzyme 2 , Endoplasmic Reticulum Stress , Mice, Inbred C57BL , Peptide Fragments , Proto-Oncogene Mas , Pyroptosis , Ventilator-Induced Lung Injury , Animals , Pyroptosis/drug effects , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/prevention & control , Endoplasmic Reticulum Stress/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin I/metabolism , Angiotensin I/pharmacology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Male , Mice , Peptidyl-Dipeptidase A/metabolism , Peptidyl-Dipeptidase A/genetics , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Humans
7.
Biomolecules ; 14(9)2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39334901

ABSTRACT

Fine particulate matter (PM2.5) is a significant cause of respiratory diseases and associated cellular damage. The mechanisms behind this damage have not been fully explained. This study investigated two types of cellular damage (inflammation and pyroptosis) induced by PM2.5, focusing on their relationship with two organelles (the endoplasmic reticulum and mitochondria). Animal models have demonstrated that PM2.5 induces excessive endoplasmic reticulum stress (ER stress), which is a significant cause of lung damage in rats. This was confirmed by pretreatment with an ER stress inhibitor (4-Phenylbutyric acid, 4-PBA). We found that, in vitro, the intracellular Ca2+ ([Ca2+]i) dysregulation induced by PM2.5 in rat alveolar macrophages was associated with ER stress. Changes in mitochondria-associated membranes (MAMs) result in abnormal mitochondrial function. This further induced the massive expression of NLRP3 and GSDMD-N, which was detrimental to cell survival. In conclusion, our findings provide valuable insights into the relationship between [Ca2+]i dysregulation, mitochondrial damage, inflammation and pyroptosis under PM2.5-induced ER stress conditions. Their interactions ultimately have an impact on respiratory health.


Subject(s)
Calcium , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Lung Injury , Mitochondria , Particulate Matter , Phenylbutyrates , Animals , Phenylbutyrates/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Calcium/metabolism , Rats , Endoplasmic Reticulum Stress/drug effects , Particulate Matter/toxicity , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Lung Injury/metabolism , Lung Injury/chemically induced , Lung Injury/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/drug effects , Male , Pyroptosis/drug effects , Rats, Sprague-Dawley , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
8.
Int J Mol Sci ; 25(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39337609

ABSTRACT

Age-related macular degeneration (AMD) is a common disease contributing to vision loss in the elderly. All-trans-retinal (atRAL) is a retinoid in the retina, and its abnormal accumulation exhibits toxicity to the retina and promotes oxidative stress-induced photoreceptor degeneration, which plays a crucial role in AMD progression. Crocin is a natural product extracted from saffron, which displays significant antioxidant and anti-inflammatory effects. The present study elucidates the protective effects of crocin on photoreceptor cell damage by atRAL and its potential mechanisms. The results revealed that crocin significantly attenuated cytotoxicity by repressing oxidative stress, mitochondrial injury, and DNA damage in atRAL-loaded photoreceptor cells. Moreover, crocin visibly inhibited DNA damage-induced apoptosis and gasdermin E (GSDME)-mediated pyroptosis in photoreceptor cells after exposure to atRAL. It was also observed that crocin distinctly prevented an increase in Fe2+ levels and lipid peroxidation caused by atRAL via suppressing the Kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor-erythroid 2-related factor 2 (NRF2)/heme oxygenase-1 (HO-1) signaling pathway, thereby ameliorating photoreceptor cell ferroptosis. In short, these findings provide new insights that crocin mitigates atRAL-induced toxicity to photoreceptor cells by inhibiting oxidative stress, apoptosis, pyroptosis, and ferroptosis.


Subject(s)
Carotenoids , Oxidative Stress , Retinaldehyde , Animals , Mice , Carotenoids/pharmacology , Oxidative Stress/drug effects , Retinaldehyde/metabolism , Cell Line , NF-E2-Related Factor 2/metabolism , Apoptosis/drug effects , Signal Transduction/drug effects , Antioxidants/pharmacology , Heme Oxygenase-1/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Ferroptosis/drug effects , DNA Damage/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Macular Degeneration/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Protective Agents/pharmacology , Lipid Peroxidation/drug effects , Pyroptosis/drug effects
9.
Int J Nanomedicine ; 19: 9175-9193, 2024.
Article in English | MEDLINE | ID: mdl-39263632

ABSTRACT

Purpose: Ischemic stroke is a refractory disease wherein the reperfusion injury caused by sudden restoration of blood supply is the main cause of increased mortality and disability. However, current therapeutic strategies for the inflammatory response induced by cerebral ischemia-reperfusion (I/R) injury are unsatisfactory. This study aimed to develop a functional nanoparticle (MM/ANPs) comprising apelin-13 (APNs) encapsulated in macrophage membranes (MM) modified with distearoyl phosphatidylethanolamine-polyethylene glycol-RVG29 (DSPE-PEG-RVG29) to achieve targeted therapy against ischemic stroke. Methods: MM were extracted from RAW264.7. PLGA was dissolved in dichloromethane, while Apelin-13 was dissolved in water, and CY5.5 was dissolved in dichloromethane. The precipitate was washed twice with ultrapure water and then resuspended in 10 mL to obtain an aqueous solution of PLGA nanoparticles. Subsequently, the cell membrane was evenly dispersed homogeneously and mixed with PLGA-COOH at a mass ratio of 1:1 for the hybrid ultrasound. DSPE-PEG-RVG29 was added and incubated for 1 h to obtain MM/ANPs. Results: In this study, we developed a functional nanoparticle delivery system (MM/ANPs) that utilizes macrophage membranes coated with DSPE-PEG-RVG29 peptide to efficiently deliver Apelin-13 to inflammatory areas using ischemic stroke therapy. MM/ANPs effectively cross the blood-brain barrier and selectively accumulate in ischemic and inflamed areas. In a mouse I/R injury model, these nanoparticles significantly improved neurological scores and reduced infarct volume. Apelin-13 is gradually released from the MM/ANPs, inhibiting NLRP3 inflammasome assembly by enhancing sirtuin 3 (SIRT3) activity, which suppresses the inflammatory response and pyroptosis. The positive regulation of SIRT3 further inhibits the NLRP3-mediated inflammation, showing the clinical potential of these nanoparticles for ischemic stroke treatment. The biocompatibility and safety of MM/ANPs were confirmed through in vitro cytotoxicity tests, blood-brain barrier permeability tests, biosafety evaluations, and blood compatibility studies. Conclusion: MM/ANPs offer a highly promising approach to achieve ischemic stroke-targeted therapy inhibiting NLRP3 inflammasome-mediated pyroptosis.


Subject(s)
Inflammasomes , Ischemic Stroke , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Nanoparticles , Pyroptosis , Animals , Mice , Ischemic Stroke/drug therapy , RAW 264.7 Cells , Pyroptosis/drug effects , Nanoparticles/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Macrophages/drug effects , Macrophages/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Male , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/chemistry , Polyethylene Glycols/chemistry , Mice, Inbred C57BL , Reperfusion Injury/drug therapy , Phosphatidylethanolamines/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism
10.
Pestic Biochem Physiol ; 204: 106030, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277358

ABSTRACT

There have been persistent concerns about the safety risks associated with DDT residues in the environment. Studies have shown that exposure to DDT or its metabolites can cause various liver diseases. However, the mechanisms of liver toxicity haven't been well studied. In our current investigation, we observed that DDT triggers pyroptosis in human liver cells (HL-7702), representing a novel form of programmed cell death. Our results delineated DDT (0-100 µM) induced pyroptosis in HL-7702 cells, which was confirmed through morphological changes, lactate dehydrogenase (LDH) release, gasdermin E (GSDME) cleavage and Annexin-V/PI staining. Knockdown of GSDME reduced cell death and transferred the mode of cell death from pyroptosis to apoptosis. Notably, DDT exposure markedly increased reactive oxygen species (ROS) production, concurrent with c-Jun N-terminal kinase (JNK) phosphorylation. Intervention with a ROS inhibitor or JNK inhibitor SP600125 restored cell viability and hindered GSDME-mediated pyroptosis. Our results firstly demonstrate that DDT suppresses HL-7702 cells growth by inducing pyroptosis mainly through the ROS/JNK/GSDME pathway. These findings not only contribute to an in-depth understanding of DDT toxicity but also open avenues for gaining valuable insights into potential mitigation strategies and therapeutic interventions.


Subject(s)
DDT , Pyroptosis , Reactive Oxygen Species , Humans , Pyroptosis/drug effects , DDT/toxicity , Reactive Oxygen Species/metabolism , Cell Line , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Apoptosis/drug effects , Gasdermins
11.
Sci Rep ; 14(1): 21860, 2024 09 19.
Article in English | MEDLINE | ID: mdl-39300102

ABSTRACT

No single treatment significantly reduces the mortality rate and improves neurological outcomes after intracerebral haemorrhage (ICH). New evidence suggests that pyroptosis-specific proteins are highly expressed in the perihaematomal tissues of patients with ICH and that the disulfiram (DSF) inhibits pyroptosis. An ICH model was established in C57BL/6 mice by intracranial injection of collagenase, after which DSF was used to treat the mice. Cell model of ICH was constructed, and DSF was used to treat the cells. HE, TUNEL, Nissl, FJC and IF staining were performed to evaluate the morphology of brain tissues; Western blotting and ELISA were performed to measure the protein expression of NOD-like receptor protein 3 (NLRP3)/Caspase-1/gasdermin D (GSDMD) classical pyroptosis pathway and Toll-likereceptor4 (TLR4)/nuclear factor-kappaB (NF-κB) inflammatory signaling pathway and blood‒brain barrier-associated factoes, and the wet/dry weight method was used to determine the brain water content. The expression of proteins related to the NLRP3/Caspase-1/GSDMD pathway and the TLR4/NF-κB pathway was upregulated in tissues surrounding the haematoma compared with that in control tissues; Moreover, the expression of the blood-brain barrier structural proteins occludin and zonula occludens-1 (ZO-1) was downregulated, and the expression of Aquaporin Protein-4 (AQP4) and matrix metalloprotein 9 (MMP-9) was upregulated. DSF significantly inhibited these changes, reduced the haematoma volume, decreased the brain water content, reduced neuronal death and degeneration and improved neurological function after ICH. ICH activated the classical pyroptosis pathway and TLR4/NF-κB inflammatory pathway, disruped the expression of blood-brain barrier structural proteins, and exacerbated brain injury and neurological dysfunction. DSF inhibited these changes and exerted the therapeutic effects on pathological changes and dysfunction caused by ICH.


Subject(s)
Blood-Brain Barrier , Disulfiram , Mice, Inbred C57BL , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Signal Transduction , Toll-Like Receptor 4 , Animals , Pyroptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Disulfiram/pharmacology , Signal Transduction/drug effects , Male , Toll-Like Receptor 4/metabolism , NF-kappa B/metabolism , Disease Models, Animal , Caspase 1/metabolism , Phosphate-Binding Proteins/metabolism , Intracranial Hemorrhages/drug therapy , Intracranial Hemorrhages/metabolism , Occludin/metabolism , Zonula Occludens-1 Protein/metabolism , Matrix Metalloproteinase 9/metabolism , Humans , Gasdermins
12.
Phytomedicine ; 134: 156021, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39255724

ABSTRACT

BACKGROUND: Cholestasis (CT) is a group of disorders caused by impaired production, secretion or excretion of bile. This may result in the deposition of bile components in the blood and liver, which in turn causes damage to liver cells and other tissues. If untreated, CT can progress to severe complications, including cirrhosis, liver failure, and potentially life-threatening conditions. OBJECTIVE: This research was intended to elucidate the function and mechanism of Paeoniflorin (PF) in ameliorating ANIT-induced pyroptosis in CT. METHODS: CT models were established in SD rats and HepG2 cells through ANIT treatment. Histological examination was conducted using haematoxylin and eosin (HE) staining to assess the histopathological alterations in the liver. Network pharmacology was employed to identify potential PF targets in CT treatment. To evaluate pyroptosis levels, various methods were used, including serum biochemical analysis, Enzyme-Linked Immunosorbent Assay (ELISA), immunofluorescence (IF), immunohistochemistry (IHC), Western blotting, transmission electron microscopy (TEM), and scanning electron microscopy (SEM). The HuProt™ 20K Chip was utilized to pinpoint potential PF-binding targets. PF's direct mechanisms in CT treatment were explored using molecular docking (MD), molecular dynamics simulations (MDS), Cellular Thermal Shift Assay (CETSA), and Surface Plasmon Resonance (SPR). RESULTS: PF administration was found to alleviate ANIT-induced liver pathology, enhance liver function markers, and improve cell viability. Network pharmacology and pyroptosis inhibitor studies suggested that PF might mitigate CT via the NLRP3-dependent pyroptosis pathway. This hypothesis was further supported by Western blotting, IF, and IHC analyses, which indicated PF's potential to inhibit NLRP3-dependent pyroptosis in CT. GSDMD was identified as a target through HuProt™ 20K Chip screening. The binding affinity of PF to GSDMD was validated through MD, MDS, CETSA, and SPR techniques. Additionally, the regulatory impact of GSDMD on downstream inflammatory pathways was confirmed by ELISA and IHC. CONCLUSION: PF exhibited a hepatoprotective effect in ANIT-induced CT, primarily by targeting GSDMD, thereby suppressing ANIT-induced pyroptosis and the subsequent release of inflammatory mediators.


Subject(s)
Cholestasis , Glucosides , Monoterpenes , Phosphate-Binding Proteins , Pyroptosis , Rats, Sprague-Dawley , Signal Transduction , Pyroptosis/drug effects , Animals , Glucosides/pharmacology , Monoterpenes/pharmacology , Humans , Signal Transduction/drug effects , Male , Rats , Hep G2 Cells , Cholestasis/drug therapy , Cholestasis/chemically induced , Phosphate-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Liver/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Molecular Docking Simulation , Disease Models, Animal , Network Pharmacology , Gasdermins
13.
Cell Mol Life Sci ; 81(1): 401, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269632

ABSTRACT

Methylglyoxal (MGO), a reactive dicarbonyl metabolite of glucose, plays a prominent role in the pathogenesis of diabetes and vascular complications. Our previous studies have shown that MGO is associated with increased oxidative stress, inflammatory responses and apoptotic cell death in endothelial cells (ECs). Pyroptosis is a novel form of inflammatory caspase-1-dependent programmed cell death that is closely associated with the activation of the NOD-like receptor 3 (NLRP3) inflammasome. Recent studies have shown that sulforaphane (SFN) can inhibit pyroptosis, but the effects and underlying mechanisms by which SFN affects MGO-induced pyroptosis in endothelial cells have not been determined. Here, we found that SFN prevented MGO-induced pyroptosis by suppressing oxidative stress and inflammation in vitro and in vivo. Our results revealed that SFN dose-dependently prevented MGO-induced HUVEC pyroptosis, inhibited pyroptosis-associated biochemical changes, and attenuated MGO-induced morphological alterations in mitochondria. SFN pretreatment significantly suppressed MGO-induced ROS production and the inflammatory response by inhibiting the NLRP3 inflammasome (NLRP3, ASC, and caspase-1) signaling pathway by activating Nrf2/HO-1 signaling. Similar results were obtained in vivo, and we demonstrated that SFN prevented MGO-induced oxidative damage, inflammation and pyroptosis by reversing the MGO-induced downregulation of the NLRP3 signaling pathway through the upregulation of Nrf2. Additionally, an Nrf2 inhibitor (ML385) noticeably attenuated the protective effects of SFN on MGO-induced pyroptosis and ROS generation by inhibiting the Nrf2/HO-1 signaling pathway, and a ROS scavenger (NAC) and a permeability transition pore inhibitor (CsA) completely reversed these effects. Moreover, NLRP3 inhibitor (MCC950) and caspase-1 inhibitor (VX765) further reduced pyroptosis in endothelial cells that were pretreated with SFN. Collectively, these findings broaden our understanding of the mechanism by which SFN inhibits pyroptosis induced by MGO and suggests important implications for the potential use of SFN in the treatment of vascular diseases.


Subject(s)
Glucose , Human Umbilical Vein Endothelial Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Pyroptosis , Pyruvaldehyde , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Pyruvaldehyde/metabolism , Pyruvaldehyde/pharmacology , Humans , Oxidative Stress/drug effects , Inflammasomes/metabolism , Inflammasomes/drug effects , Animals , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Glucose/metabolism , Isothiocyanates/pharmacology , Mice , Sulfoxides/pharmacology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Male , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
14.
BMC Res Notes ; 17(1): 260, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267194

ABSTRACT

This study hypothesized that SCFA, acetate impacts positively on hypothalamic pyroptosis and its related abnormalities in experimentally induced PCOS rat model, possibly through NrF2/HIF1-α modulation. Eight-week-old female Wister rats were divided into groups (n = 5), namely control, PCOS, acetate and PCOS + acetate groups. Induction of PCOS was performed by administering 1 mg/kg body weight of letrozole for 21 days. After PCOS confirmation, the animals were treated with 200 mg/kg of acetate for 6 weeks. Rats with PCOS were characterized with insulin resistance, leptin resistance, increased plasma testosterone as well as degenerated ovarian follicles. There was also a significant increase in hypothalamic triglyceride level, triglyceride-glucose index, inflammatory biomarkers (SDF-1 and NF-kB) and caspase-6 as well as plasma LH and triglyceride. A decrease was observed in plasma adiponectin, GnRH, FSH, and hypothalamic GABA with severe inflammasome expression in PCOS rats. These were accompanied by decreased level of NrF2/HIF1-α, and the alterations were reversed when treated with acetate. Collectively, the present results suggest the therapeutic impact of acetate on hypothalamic pyroptosis and its related comorbidity in PCOS, a beneficial effect that is accompanied by modulation of NrF2/HIF1-α.


Subject(s)
Hypothalamus , Hypoxia-Inducible Factor 1, alpha Subunit , Polycystic Ovary Syndrome , Pyroptosis , Rats, Wistar , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Female , Animals , Hypothalamus/metabolism , Hypothalamus/drug effects , Hypothalamus/pathology , Pyroptosis/drug effects , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance , NF-E2-Related Factor 2/metabolism , Disease Models, Animal , Letrozole/pharmacology , Triglycerides/blood , Triglycerides/metabolism , Luteinizing Hormone/blood , Follicle Stimulating Hormone/blood , Adiponectin/metabolism , Adiponectin/blood , Testosterone/blood , Leptin/blood , Leptin/metabolism , Gonadotropin-Releasing Hormone/metabolism , gamma-Aminobutyric Acid/metabolism
15.
Cell Death Dis ; 15(9): 662, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256356

ABSTRACT

Ovarian cancer, the second most leading cause of gynecologic cancer mortality worldwide, is challenged by chemotherapy resistance, presenting a significant hurdle. Pyroptosis, an inflammation-linked programmed cell death mediated by gasdermins, has been shown to impact chemoresistance when dysregulated. However, the mechanisms connecting pyroptosis to chemotherapy resistance in ovarian cancer are unclear. We found that cytokine receptor-like factor 1 (CRLF1) is a novel component of mTORC2, enhancing AKT Ser473 phosphorylation through strengthening the interaction between AKT and stress-activated protein kinase interacting protein 1 (SIN1), which in turn inhibits the mitogen-activated protein kinase kinase kinase 5 (ASK1)-JNK-caspase-3-gasdermin E pyroptotic pathway and ultimately confers chemoresistance. High CRLF1-expressing tumors showed sensitivity to AKT inhibition but tolerance to cisplatin. Remarkably, overexpression of binding-defective CRLF1 variants impaired AKT-SIN1 interaction, promoting pyroptosis and chemosensitization. Thus, CRLF1 critically regulates chemoresistance in ovarian cancer by modulating AKT/SIN1-dependent pyroptosis. Binding-defective CRLF1 variants could be developed as tumor-specific polypeptide drugs to enhance chemotherapy for ovarian cancer.


Subject(s)
Adaptor Proteins, Signal Transducing , Drug Resistance, Neoplasm , Mechanistic Target of Rapamycin Complex 2 , Ovarian Neoplasms , Proto-Oncogene Proteins c-akt , Pyroptosis , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Drug Resistance, Neoplasm/drug effects , Pyroptosis/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Cell Line, Tumor , Animals , Mice , Cisplatin/pharmacology , Cisplatin/therapeutic use , Mice, Nude , Signal Transduction/drug effects
16.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(9): 967-973, 2024.
Article in Chinese | MEDLINE | ID: mdl-39267513

ABSTRACT

OBJECTIVES: To study the effects and mechanisms of tetramethylpyrazine (TMP) on tumor necrosis factor-α (TNF-α)-induced inflammatory injury in human coronary artery endothelial cells (HCAEC). METHODS: HCAEC were randomly divided into four groups: the control group (no treatment), the model group (treated with TNF-α, 50 ng/mL for 24 hours), the TMP group (pre-treated with TMP, 80 µg/mL for 12 hours followed by TNF-α treatment for 24 hours), and the SIRT1 inhibitor group (pre-treated with TMP and the specific SIRT1 inhibitor EX527 for 12 hours followed by TNF-α treatment for 24 hours). Cell viability was assessed using the CCK-8 method, lactate dehydrogenase (LDH) activity was measured using an LDH assay kit, reactive oxygen species (ROS) levels were observed using DCFH-DA staining, expression of pyroptosis-related proteins was detected by Western blot, and SIRT1 expression was analyzed using immunofluorescence staining. RESULTS: Compared to the control group, the model group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). Compared to the model group, the TMP group exhibited increased cell viability, decreased LDH activity, ROS level and expression of pyroptosis-related proteins, and increased SIRT1 expression (P<0.05). In comparison to the TMP group, the SIRT1 inhibitor group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). CONCLUSIONS: TMP may attenuate TNF-α-induced inflammatory injury in HCAEC, which is associated with the inhibition of pyroptosis and activation of the SIRT1 signaling pathway.


Subject(s)
Endothelial Cells , Pyrazines , Reactive Oxygen Species , Signal Transduction , Sirtuin 1 , Tumor Necrosis Factor-alpha , Sirtuin 1/metabolism , Sirtuin 1/physiology , Humans , Pyrazines/pharmacology , Signal Transduction/drug effects , Endothelial Cells/drug effects , Tumor Necrosis Factor-alpha/metabolism , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Pyroptosis/drug effects , Cells, Cultured , Inflammation/drug therapy
17.
Cell Death Dis ; 15(9): 695, 2024 Sep 29.
Article in English | MEDLINE | ID: mdl-39343834

ABSTRACT

Chemotherapy including platinum-based drugs are a possible strategy to enhance the immune response in advanced melanoma patients who are resistant to immune checkpoint blockade (ICB) therapy. However, the immune-boosting effects of these drugs are a subject of controversy, and their impact on the tumor microenvironment are poorly understood. In this study, we discovered that lipid peroxidation (LPO) promotes the formation of lipid rafts in the membrane, which mediated by Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4) impairs the sensitivity of melanoma cells to platinum-based drugs. This reduction primarily occurs through the inhibition of immunogenic ferroptosis and pyroptosis by reducing cell membrane pore formation. By disrupting ACSL4-mediaged lipid rafts via the removal of membrane cholesterol, we promoted immunogenic cell death, transformed the immunosuppressive environment, and improved the antitumor effectiveness of platinum-based drugs and immune response. This disruption also helped reverse the decrease in CD8+ T cells while maintaining their ability to secrete cytokines. Our results reveal that ACSL4-dependent LPO is a key regulator of lipid rafts formation and antitumor immunity, and that disrupting lipid rafts has the potential to enhance platinum-based drug-induced immunogenic ferroptosis and pyroptosis in melanoma. This novel strategy may augment the antitumor immunity of platinum-based therapy and further complement ICB therapy.


Subject(s)
Coenzyme A Ligases , Immunogenic Cell Death , Melanoma , Membrane Microdomains , Membrane Microdomains/metabolism , Membrane Microdomains/drug effects , Coenzyme A Ligases/metabolism , Melanoma/pathology , Melanoma/drug therapy , Melanoma/immunology , Animals , Humans , Mice , Immunogenic Cell Death/drug effects , Cell Line, Tumor , Ferroptosis/drug effects , Mice, Inbred C57BL , Lipid Peroxidation/drug effects , Pyroptosis/drug effects , Tumor Microenvironment/drug effects
18.
J Zhejiang Univ Sci B ; 25(9): 756-772, 2024 Sep 12.
Article in English, Chinese | MEDLINE | ID: mdl-39308066

ABSTRACT

Doxorubicin (DOX) is a commonly administered chemotherapy drug for treating hematological malignancies and solid tumors; however, its clinical application is limited by significant cardiotoxicity. Cynaroside (Cyn) is a flavonoid glycoside distributed in honeysuckle, with confirmed potential biological functions in regulating inflammation, pyroptosis, and oxidative stress. Herein, the effects of Cyn were evaluated in a DOX-induced cardiotoxicity (DIC) mouse model, which was established by intraperitoneal injections of DOX (5 mg/kg) once a week for three weeks. The mice in the treatment group received dexrazoxane, MCC950, and Cyn every two days. Blood biochemistry, histopathology, immunohistochemistry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and western blotting were conducted to investigate the cardioprotective effects and potential mechanisms of Cyn treatment. The results demonstrated the significant benefits of Cyn treatment in mitigating DIC; it could effectively alleviate oxidative stress to a certain extent, maintain the equilibrium of cell apoptosis, and enhance the cardiac function of mice. These effects were realized via regulating the transcription levels of pyroptosis-related genes, such as nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), caspase-1, and gasdermin D (GSDMD). Mechanistically, for DOX-induced myocardial injury, Cyn could significantly modulate the expression of pivotal genes, including adenosine monophosphate-activated protein kinase (AMPK), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), sirtuin 3 (SIRT3), and nuclear factor erythroid 2-related factor 2 (Nrf2). We attribute it to the mediation of AMPK/SIRT3/Nrf2 pathway, which plays a central role in preventing DOX-induced cardiomyocyte injury. In conclusion, the present study confirms the therapeutic potential of Cyn in DIC by regulating the AMPK/SIRT3/Nrf2 pathway.


Subject(s)
AMP-Activated Protein Kinases , Cardiotoxicity , Doxorubicin , Myocytes, Cardiac , NF-E2-Related Factor 2 , Pyroptosis , Signal Transduction , Sirtuin 3 , Animals , Doxorubicin/adverse effects , Pyroptosis/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Mice , NF-E2-Related Factor 2/metabolism , AMP-Activated Protein Kinases/metabolism , Sirtuin 3/metabolism , Cardiotoxicity/prevention & control , Cardiotoxicity/drug therapy , Male , Signal Transduction/drug effects , Oxidative Stress/drug effects , Mice, Inbred C57BL
19.
J Cell Mol Med ; 28(18): e70112, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39320020

ABSTRACT

Oral submucous fibrosis (OSF) is a precancerous condition in the oral cavity, which is closely related to the myofibroblast conversion of buccal mucosal fibroblasts (BMFs) after chronic consumption of areca nut. Emerging evidence suggests pyroptosis, a form of programmed cell death that is mediated by inflammasome, is implicated in persistent myofibroblast activation and fibrosis. Besides, numerous studies have demonstrated the effects of non-coding RNAs on pyroptosis and myofibroblast activities. Herein, we aimed to target key long non-coding RNA PVT1 with natural compound, carvacrol, to alleviate pyroptosis and myofibroblast activation in OSF. We first identified PVT1 was downregulated in the carvacrol-treated fBMFs and then demonstrated that myofibroblast features and expression of pyroptosis makers were all reduced in response to carvacrol treatment. Subsequently, we analysed the expression of PVT1 and found that PVT1 was aberrantly upregulated in OSF specimens and positively correlated with several fibrosis markers. After revealing the suppressive effects of carvacrol on myofibroblast characterisitcs and pyroptosis were mediated by repression of PVT1, we then explored the potential mechanisms. Our data showed that PVT1 may serve as a sponge of microRNA(miR)-20a to mitigate the myofibroblast activation and pyroptosis. Altogether, these findings indicated that the anti-fibrosis effects of carvacrol merit consideration and may be due to the attenuation of pyroptosis and myofibroblast activation by targeting the PVT1/miR-20a axis.


Subject(s)
Cymenes , MicroRNAs , Myofibroblasts , Oral Submucous Fibrosis , Pyroptosis , RNA, Long Noncoding , Oral Submucous Fibrosis/pathology , Oral Submucous Fibrosis/genetics , Oral Submucous Fibrosis/metabolism , Oral Submucous Fibrosis/drug therapy , Pyroptosis/drug effects , Pyroptosis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Cymenes/pharmacology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Myofibroblasts/pathology , Disease Progression , Down-Regulation/drug effects , Fibroblasts/metabolism , Fibroblasts/drug effects
20.
Int Immunopharmacol ; 141: 112990, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39223062

ABSTRACT

Inflammatory bowel diseases (IBDs) are chronic, relapsing, and inflammatory disorders of the gastrointestinal tract characterized by abnormal immune responses. Recently, STING has emerged as a promising therapeutic target for various autoinflammatory diseases. However, few STING-selective small molecules have been investigated as novel strategies for IBD. In this study, we sought to examine the effects of PROTAC-based STING degrader SP23 on acute colitis and explore its underlying mechanism. SP23 treatment notably alleviates dextran sulfate sodium (DSS)-induced colitis. Pharmacological degradation of STING significantly reduced the production of inflammatory cytokines, such as TNF-α, IL-1ß, and IL-6, and inhibited macrophage polarization towards the M1 type. Furthermore, SP23 administration decreased the loss of tight junction proteins, including ZO-1, occludin, and claudin-1, and downregulated STING and NLRP3 signaling pathways in intestinal inflammation. In vitro, STING activated NLRP3 inflammasome-mediated pyroptosis in intestinal epithelial cells, which could be abrogated by SP23 and STING siRNA intervention. In conclusion, these findings provide new evidence for STING as a novel therapeutic target for IBD, and reveal that hyperactivation of STING could exaggerate colitis by inducing NLRP3/Caspase-1/GSDMD axis mediated intestinal epithelial cells pyroptosis.


Subject(s)
Colitis , Dextran Sulfate , Macrophages , Membrane Proteins , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Pyroptosis/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Colitis/drug therapy , Colitis/chemically induced , Colitis/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Signal Transduction/drug effects , Inflammasomes/metabolism , Cytokines/metabolism , Male , Humans , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/immunology , Disease Models, Animal , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL