Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Phytother Res ; 38(1): 82-97, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37807970

ABSTRACT

Ursolic acid (UA) is a naturally occurring pentacyclic triterpenoid widely found in fruits and vegetables. It has been reported that UA has anti-inflammatory effects. However, its efficacy and mechanism of action in the treatment of chronic prostatitis (CP) remain unclear. This study aimed to investigate the efficacy of UA treatment in CP and further explore the underlying mechanism. CP rat and pyroptosis cell models were established in vivo and in vitro, respectively. The efficacy of UA in inhibiting CP was evaluated via haematoxylin-eosin (HE) staining and measurement of inflammatory cytokines. RNA sequencing and molecular docking were used to predict the therapeutic targets of UA in CP. The expression of pyroptosis-related proteins was examined using various techniques, including immunohistochemistry, immunofluorescence, and flow cytometry. UA significantly ameliorated pathological damage and reduced the levels of proinflammatory cytokines in the CP model rats. RNA sequencing analysis and molecular docking suggested that NLRP3, Caspase-1, and GSDMD may be key targets. We also found that UA decreased ROS levels, alleviated oxidative stress, and inhibited p-NF-κB protein expression both in vivo and in vitro. UA improved pyroptosis morphology as indicated by electron microscope and inhibited the expression of the pyroptosis-related proteins NLRP3, Caspase-1, ASC, and GSDMD, reversed the levels of IL-1ß, IL-18, and lactate dehydrogenase in vivo and in vitro. UA can mitigate CP by regulating the NLRP3 inflammasome-mediated Caspase-1/GSDMD pathway. Therefore, UA may be a potential for the treatment of CP.


Subject(s)
Inflammasomes , Prostatitis , Humans , Male , Rats , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Ursolic Acid , Pyroptosis/physiology , Caspase 1/metabolism , Prostatitis/drug therapy , Molecular Docking Simulation , Gasdermins , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology
2.
Gene ; 893: 147888, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-37839766

ABSTRACT

BACKGROUND: Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), acting as one common sepsis-associated organ injury, induces uncontrolled and self-amplifies pulmonary inflammation. Given the lack of clinically effective approaches, the mortality rate of it still remains high. Suramin(SUR), as an antiparasitic drug initially, was found to ameliorate sepsis associated ALI in our previous work. However, the underlying mechanism of its protective effects has not been clarified. Pyroptosis, categorized as an inflammatory form of programmed cell death, could aggravate lung inflammatory responses via inducing alveolar macrophages (AM) pyroptosis. METHODS: MH-S AM cell line was stimulated with or without lipopolysaccharide (LPS) or suramin, and the differential expression genes (DEGs) were excavated using RNA sequencing (RNA-seq). To identify the regulatory roles of these genes, pyroptosis-related genes (PRGs), GO/KEGG and GSEA analysis were conducted. We also performed WB, qRTPCR and ELISA to validate the RNA-seq results and further expound the protective effect of suramin. RESULTS: 624 DEGs were identified between control (CON) and lipopolysaccharide (LPS) groups, and enrichment analysis of these genes revealed significantly enriched pathways that related to immune system and signal transduction. Meanwhile, 500 DEGs were identified in LPS/SUR+LPS group. In addition to the pathways mentioned above, IL-17 pathway and C-type lectin receptor signaling pathway were also enriched. All 6 pathways were connected with pyroptosis. Concurrently, the "DESeq2" R package was used to identify differentially expressed PRGs. Nod1, Nod2, interleukin (IL)-1b, IL-6, tumor necrosis factor (TNF), NLRP3 were upregulated under LPS stimulation. Then, in SUR+LPS group, Nod2, IL-6, IL-1b, NLRP3 were downregulated. The validation results of WB, qRT-PCR, and ELISA showed: the protein and mRNA expression levels of NLRP3, caspase-1, GSDMD and the concentrations of IL-1b, IL-18 were decreased when treated with suramin and LPS. CONCLUSION: Suramin could inhibit NLRP3/caspase-1/GSDMD canonical pyroptosis pathway in LPS-induced MH-S alveolar macrophages.


Subject(s)
Macrophages, Alveolar , Sepsis , Humans , Macrophages, Alveolar/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Lipopolysaccharides/pharmacology , Suramin/pharmacology , Interleukin-6/genetics , RNA-Seq , Inflammasomes/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/pharmacology , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/pharmacology
3.
Cell Mol Biol (Noisy-le-grand) ; 69(10): 227-232, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37953558

ABSTRACT

Traumatic arthritis is caused by mechanical injury and results in the degeneration of articular cartilage, but it is unclear whether it is related to the pyroptosis of chondrocyte (CHs). Thus, this study was designed to investigate the role of GSDMD, the executor of pyroptosis, in the human cartilage during mechanical injury. We collected the human hip joint and used a loading apparatus to produce compression on the cartilage disc. After one hour of 15 MPa or 25 MPa injury, the acute and chronic effects of the mechanical injury on the cartilage were tested. We stained the CHs in the cartilage with calcein and DAPI to calculate the live-cell rate. The chondrogenic phenotype was determined by analyzing the mRNA levels of type II collagen alpha 1 (Col2A1), type I collagen alpha 2 (Col2A1), and SOX9. Besides, the pyroptosis process was determined by the mRNA levels of caspase-1/5, GSDMD, IL-1ß, and IL-18. We also explored the preventive role and therapeutic role of GSDMD inhibitors in mechanical injury via culturing the cartilage before and after the compression, respectively. Mechanical compression injured the viability and function of CHs in cartilage partly based on the pyroptosis. The pretreatment of GSDMD inhibitor in cartilage before injury could maintain the live cells and Col2A1 expression and prevent pyroptosis after injury. Besides, supplying the cartilage with GSDMD inhibitor after injury also alleviated the cell death and dysfunction of CHs, and suppressed the pyroptosis. Using an inhibitor of GSDMD can play a preventive role and play a therapeutic role in the mechanical injury of cartilage.


Subject(s)
Cartilage, Articular , Chondrocytes , Gasdermins , Phosphate-Binding Proteins , Humans , Cartilage, Articular/metabolism , Caspases/metabolism , Chondrocytes/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Pyroptosis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gasdermins/antagonists & inhibitors
4.
Curr Pharm Des ; 29(42): 3408-3420, 2023.
Article in English | MEDLINE | ID: mdl-37936452

ABSTRACT

INTRODUCTION: Breast cancer has emerged as the most widespread cancer globally surpassing lung cancer, and has become a primary cause of mortality among women. While MFHAS1 has been implicated in the pathophysiology of various diseases, its precise involvement in breast cancer remains unclear. METHODS: This study endeavors to elucidate the regulatory function of MFHAS1 in breast cancer cell pyroptosis and the associated molecular mechanisms. Our findings indicate that the inhibition of MFHAS1 can impede the proliferation and invasion of breast cancer cells, while also inducing cell pyroptosis via caspase1-dependent activation of GSDMD. RESULTS: This process results in the cleavage of cell membranes, leading to the release of inflammatory factors and LDH. Subsequent investigations revealed that the silencing of MFHAS1 can promote JNK phosphorylation, thereby activating the JNK signaling cascade. Notably, this effect can be counteracted by the JNK-specific inhibitor sp600125. Ultimately, our investigation substantiated the identical function of MFHAS1 in breast cancer tissue derived from animal models. CONCLUSION: To summarize, our findings demonstrate that the inhibition of MFHAS1 elicits pyroptosis in human breast cancer cells through the facilitation of JNK phosphorylation and the activation of the downstream NF-κB/caspase-1/GSDMD signaling cascade, thereby proposing the prospect of MFHAS1 as a viable therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms , Pyroptosis , Animals , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Gasdermins , MAP Kinase Signaling System , NF-kappa B/metabolism , Oncogene Proteins/metabolism , Oncogene Proteins/pharmacology , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Pyroptosis/genetics , Pyroptosis/physiology , Signal Transduction , Caspase 1/metabolism
5.
J Exp Clin Cancer Res ; 42(1): 274, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37864196

ABSTRACT

BACKGROUND: Tumor cells can resist chemotherapy-induced pyroptosis through glycolytic reprogramming. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. Herein, we refine the oncogenic role of ERRα in the pyroptosis pathway and glycolytic metabolism. METHODS: The interaction between ERRα and HIF-1α was verified using co-immunoprecipitation. The transcriptional binding sites of ERRα and NLRP3 were confirmed using dual-luciferase reporter assay and cleavage under targets and tagmentation (CUT&Tag). Flow cytometry, transmission electron microscopy, scanning electron microscopy, cell mito stress test, and extracellular acidification rate analysis were performed to investigate the effects of ERRα on the pyroptosis pathway and glycolytic metabolism. The results of these experiments were further confirmed in endometrial cancer (EC)-derived organoids and nude mice. In addition, the expression of ERRα-related pyroptosis genes was analyzed using The Cancer Genome Atlas and Gene Expression Omnibus database. RESULTS: Triggered by a hypoxic microenvironment, highly expressed ERRα could bind to the promoter of NLRP3 and inhibit caspase-1/GSDMD signaling, which reduced inflammasome activation and increased pyroptosis resistance, thereby resulting in the resistance of cancer cells to cisplatin. Moreover, ERRα activated glycolytic rate-limiting enzyme to bridge glycolytic metabolism and pyroptosis in EC. This phenomenon was further confirmed in EC-derived organoids and nude mice. CUT & Tag sequencing and The Cancer Genome Atlas database analysis showed that ERRα participated in glycolysis and programmed cell death, which resulted in EC progression. CONCLUSIONS: ERRα inhibits pyroptosis in an NLRP3-dependent manner and induces glycolytic metabolism, resulting in cisplatin resistance in EC cells.


Subject(s)
Endometrial Neoplasms , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Mice , Animals , Female , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Caspase 1/pharmacology , Mice, Nude , Pyroptosis , Cisplatin/pharmacology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Glycolysis , Tumor Microenvironment , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Pore Forming Cytotoxic Proteins/metabolism , ERRalpha Estrogen-Related Receptor
6.
DNA Cell Biol ; 42(9): 554-562, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37566540

ABSTRACT

Inhibition of the inflammatory response triggered by microglial pyroptosis inflammatory activation may be one of the effective ways to alleviate cerebral ischemia-reperfusion injury, the specific mechanism of which remains unclear. In this study, BV-2 microglia with or without oxygen-glucose deprivation/reoxygenation (OGD/R) or long noncoding RNA (lncRNA) Gm44206 knockdown were used as cell models to conduct an in vitro study. Detection of lactate dehydrogenase release and pyroptosis-related protein levels was performed using a corresponding kit and western blotting, respectively. Proliferation of microglia was evaluated by CCK8 assay. Enzyme-linked immunosorbent assay was applied for measuring levels of proinflammatory cytokines. This study verified the involvement of microglial pyroptosis as well as upregulation of NLRP3, Caspase-1, GSDMD, and Apoptosis-associated Speck-like protein containing a C-terminal caspase-recruitment domain (ASC) in cerebral ischemia-reperfusion injury. Moreover, knockdown of lncRNA Gm44206 could alleviate OGD/R-induced microglial pyroptosis and cell proliferation inhibition through the NLRP3/Caspase-1/GSDMD pathway, thus decreasing the release of proinflammatory cytokines, including interleukin (IL)-1ß, IL-6, IL-18, and tumor necrosis factor-alpha. In conclusion, this study established a correlation between microglial pyroptosis and cerebral ischemia-reperfusion injury and identified lncRNA Gm44206 as a potential regulator of NLRP3/Caspase-1/GSDMD axis-mediated microglial pyroptosis, which could be considered a promising therapeutic target.


Subject(s)
RNA, Long Noncoding , Reperfusion Injury , Humans , Pyroptosis/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Microglia/pathology , Caspase 1/genetics , Caspase 1/metabolism , Reperfusion Injury/metabolism , Interleukin-18/metabolism , Interleukin-18/pharmacology , Oxygen/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Pore Forming Cytotoxic Proteins/metabolism
7.
J Biochem Mol Toxicol ; 37(8): e23444, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37393521

ABSTRACT

Saikosaponin-D (SSD), an active ingredient in Bupleurum chinense, exerts anticancer effects in various cancers by inhibiting cancer proliferation and inducing apoptosis. However, whether SSD can induce other forms of cell death is unknown. The current study aims to demonstrate that SSD can induce pyroptosis in non-small-cell lung cancer. In this study, HCC827 and A549 non-small-cell lung cancer cells were treated with different concentrations of SSD for 1.5 h. HE and TUNEL staining were used to verify cell damage caused by SSD. Immunofluorescence and western blotting were performed to verify the effect of SSD on the NF-κB/NLRP3/caspase-1/gasdermin D (GSDMD) pathway. Changes in inflammatory factors were detected by ELISAs. Finally, the reactive oxygen species (ROS) scavenger N-acetylcysteine (NAC) was introduced to verify that SSD induces pyroptosis through the ROS/NF-κB pathway. The results of the HE and TUNEL staining showed that SSD resulted in balloon-like swelling of NSCLC cells accompanied by increased DNA damage. Immunofluorescence and western blot assays confirmed that SSD treatment activated the NLRP3/caspase-1/GSDMD pathway, stimulated an increase in ROS levels and activated NF-κB in lung cancer cells. The ROS scavenger N-acetylcysteine significantly attenuated SSD-induced NF-κB/NLRP3/caspase-1/GSDMD pathway activation and inhibited the release of the inflammatory cytokines IL-1ß and IL-18. In conclusion, SSD induced lung cancer cell pyroptosis by inducing ROS accumulation and activating the NF-κB/NLRP3/caspase-1/GSDMD pathway. These experiments lay the foundation for the application of SSD in the treatment of non-small-cell lung cancer and regulation of the lung cancer immune microenvironment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Pyroptosis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase 1/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Acetylcysteine/pharmacology , Lung Neoplasms/drug therapy , Inflammasomes/metabolism , Tumor Microenvironment , Phosphate-Binding Proteins/pharmacology , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/pharmacology
8.
Chin J Nat Med ; 21(6): 423-435, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37407173

ABSTRACT

Acute lung injury (ALI) is a prevalent and severe clinical condition characterized by inflammatory damage to the lung endothelial and epithelial barriers, resulting in high incidence and mortality rates. Currently, there is a lack of safe and effective drugs for the treatment of ALI. In a previous clinical study, we observed that Jinyinqingre oral liquid (JYQR), a Traditional Chinese Medicine formulation prepared by the Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, exhibited notable efficacy in treating inflammation-related hepatitis and cholecystitis in clinical settings. However, the potential role of JYQR in ALI/acute respiratory distress syndrome (ARDS) and its anti-inflammatory mechanism remains unexplored. Thus, the present study aimed to investigate the therapeutic effects and underlying molecular mechanisms of JYQR in ALI using a mouse model of lipopolysaccharide (LPS)-induced ALI and an in vitro RAW264.7 cell model. JYQR yielded substantial improvements in LPS-induced histological alterations in lung tissues. Additionally, JYQR administration led to a noteworthy reduction in total protein levels within the BALF, a decrease in MPAP, and attenuation of pleural thickness. These findings collectively highlight the remarkable efficacy of JYQR in mitigating the deleterious effects of LPS-induced ALI. Mechanistic investigations revealed that JYQR pretreatment significantly inhibited NF-κB activation and downregulated the expressions of the downstream proteins, namely NLRP3 and GSDMD, as well as proinflammatory cytokine levels in mice and RAW2647 cells. Consequently, JYQR alleviated LPS-induced ALI by inhibiting the NF-κB/NLRP3/GSDMD pathway. JYQR exerts a protective effect against LPS-induced ALI in mice, and its mechanism of action involves the downregulation of the NF-κB/NLRP3/GSDMD inflammatory pathway.


Subject(s)
Acute Lung Injury , NF-kappa B , Humans , NF-kappa B/metabolism , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Lung , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Phosphate-Binding Proteins/therapeutic use , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/pharmacology , Pore Forming Cytotoxic Proteins/therapeutic use
9.
BMC Mol Cell Biol ; 23(1): 54, 2022 Dec 14.
Article in English | MEDLINE | ID: mdl-36517743

ABSTRACT

BACKGROUND: Pyroptosis of endothelial cells is a new cause of endothelial dysfunction in multiple diseases. Ceramide acts as a potential bioactive mediator of inflammation and increases vascular endothelial permeability in many diseases, whether it can aggravate vascular endothelial injury by inducing cell pyroptosis remains unknown. This study was established to explore the effects of C8-ceramide (C8-Cer) on human umbilical vein vascular endothelial cells (HUVECs) and its possible underlying mechanism. METHODS: HUVECs were exposed to various concentrations of C8-Cer for 12 h, 24 h, 48 h. The cell survival rate was measured using the cell counting kit-8 assay. Western blotting and Real-time polymerase chain reaction (RT-PCR) were used to detect the pyroptosis-releated protein and mRNA expressions, respectively. Caspase-1 activity assay was used to detect caspase-1 activity. Hoechst 33342/propidium iodide double staining and flow cytometry were adopted to measure positive staining of cells. Lactate dehydrogenase release assay and enzyme-linked immunosorbent assay were adopted to measure leakage of cellular contents. FITC method was used to detect the permeability of endothelial cells. ROS fluorescence intensity were detected by flow cytometry. RESULTS: The viability of HUVECs decreased gradually with the increase in ceramide concentration and time. Ceramide upregulated the expression of thioredoxin interacting protein (TXNIP), NLRP3, GSDMD, GSDMD-NT, caspase-1 and Casp1 p20 at the protein and mRNA level in a dose-dependent manner. It also enhanced the PI uptake in HUVECs and upregulated caspase-1 activity. Moreover, it promoted the release of lactate dehydrogenase, interleukin-1ß, and interleukin-18. Meanwhile, we found that ceramide led to increased vascular permeability. The inhibitor of NLRP3 inflammasome assembly, MCC950, was able to disrupt the aforementioned positive loop, thus alleviating vascular endothelial cell damage. Interestingly, inhibition of TXNIP either chemically using verapamil or genetically using small interfering RNA (siRNA) can effectively inhibit ceramide-induced pyroptosis and improved cell permeability. In addition, ceramide stimulated reactive oxygen species (ROS) generation. The pretreatment of antioxidant N-acetylcysteine (NAC), ROS scavenger, blocked the expression of pyroptosis markers induced by C8-cer in HUVECs. CONCLUSION: The current study demonstrated that C8-Cer could aggravate vascular endothelial cell damage and increased cell permeability by inducing cell pyroptosis. The results documented that the ROS-dependent TXNIP/NLRP3/GSDMD signalling pathway plays an essential role in the ceramide-induced pyroptosis in HUVECs.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Humans , Pyroptosis/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Ceramides/pharmacology , Caspase 1/genetics , Caspase 1/metabolism , Caspase 1/pharmacology , Human Umbilical Vein Endothelial Cells , RNA, Messenger/genetics , Lactate Dehydrogenases/metabolism , Carrier Proteins , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Pore Forming Cytotoxic Proteins/metabolism
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(9): 1171-1181, 2022 Sep 28.
Article in English, Chinese | MEDLINE | ID: mdl-36411700

ABSTRACT

OBJECTIVES: Trimethylamine oxide (TMAO) is a metabolite of intestinal flora and is known to promote the progression of atherosclerotic plaques. However, how TMAO works, including its effect on vascular endothelial cells, is not fully understood. This study aims to explore the biological role of TMAO in human umbilical vein endothelial cells (HUVECs) and the underlying mechanism. METHODS: Cell pyroptosis and the loss of plasma membrane integrity were induced under TMAO stimulation in HUVECs. The plasma membrane integrity of the cells was measured by Hoechst 33342/propidium iodide (PI) staining and lactate dehydrogenase leakage assay, and the changes in cell morphology were observed by atomic force microscope. The expression of proteins related to pyroptosis was determined by Western blotting or immunofluorescence. Mitochondrial acetaldehyde dehydrogenase 2 (ALDH2) activity in HUVECs was measured by the ALDH2 activity assay kit, and the level of reactive oxygen species (ROS) was detected by fluorescent probe DCFH-DA. RESULTS: TMAO induced pyroptotic cell death, manifesting by the presence of propidium iodide-positive cells, the leakage of lactate dehydrogenase, the production of N-terminal gasdermin D (GSDMD-N), and the formation of plasma membrane pores. Moreover, TMAO induced elevated expression of inflammasome components, nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), and caspase-1 in cells. TMAO significantly inhibited ALDH2 activity and increased intracellular ROS production. However, the activation of ALDH2 by pharmacological manipulation attenuated TMAO-induced inflammasome activation and GSDMD-N production. CONCLUSIONS: TMAO induces pyroptosis of vascular endothelial cells through the ALDH2/ROS/NLRP3/GSDMD signaling pathway, which may be a potential therapeutic target for improving the treatment of atherosclerosis.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Humans , Inflammasomes/metabolism , Reactive Oxygen Species , Propidium/pharmacology , Human Umbilical Vein Endothelial Cells , Lactate Dehydrogenases/metabolism , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism
11.
Oncogene ; 41(47): 5092-5106, 2022 11.
Article in English | MEDLINE | ID: mdl-36245058

ABSTRACT

Gasdermin D (GSDMD) has recently been identified as a cytoplasmic effector protein that plays a central role in pyroptosis of immune cells. However, GSDMD is a universally expressed protein, and its function beyond pyroptosis, especially in cancer cells, has not been well characterized. Here, we report that predominant localization of GSDMD in the nucleoplasm in vivo indicates favorable clinical outcomes in colorectal cancer, while a lack of nuclear localization of GSDMD is associated with poor outcomes. Nuclear GSDMD, rather than cytoplasmic GSDMD, inhibits cell growth and promotes apoptosis in colorectal cancer. Hypoxia in the tumor microenvironment accounts for mild or moderate nuclear translocation of GSDMD in vivo. Under the stimulation of chemotherapy drugs, nuclear GSDMD promotes apoptosis via regulation of its subcellular distribution rather than pyroptosis-related cleavage. After nuclear translocation, GSDMD interacts with PARP-1 to dramatically inhibit its DNA damage repair-related function by functioning like the PARP inhibitor olaparib, thus forming a "hypoxia/chemotherapy-GSDMD nuclear translocation-PARP-1 blockade-DNA damage and apoptosis" axis. This study redefines the pyroptosis-independent function of GSDMD and suggests that the subcellular localization of GSDMD may serve as a molecular indicator of clinical outcomes and a promising therapeutic target in colorectal cancer.


Subject(s)
Colorectal Neoplasms , Pyroptosis , Humans , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Hypoxia , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Tumor Microenvironment
12.
Med Sci Monit ; 27: e928411, 2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33690262

ABSTRACT

BACKGROUND The inflammation and apoptosis of podocytes contribute to the pathological progression of diabetic nephropathy. Gasdermin D (GSDMD) plays an executive role in pyroptosis, but its effect on high-glucose (HG)-induced inflammation and apoptosis remains unclear. The aim of this study was to investigate the effect of GSDMD on high-glucose-induced inflammation and apoptosis in podocytes. MATERIAL AND METHODS Mouse podocytes were cultivated by high- or normal-glucose medium. We used western blot analysis, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and immunofluorescence to detect the expression and localization of GSDMD in high-glucose-induced podocytes, and the expression of apoptosis-related proteins Bax and Bcl-2, inflammatory factors IL-1ß, IL-6, and TNF-alpha, and JNK pathways in high-glucose-induced podocytes. Western blot and immunofluorescence were used to detect the expression and localization of synaptopodin under GSDMD knockdown and JNK-specific blocker SP600125. MitoSOX Red was used to detect the production of ROS in mitochondria under siGSDMD. The intracellular ROS generation was detected using a reactive oxygen species assay kit. RESULTS We found that GSDMD knockdown and JNK inhibition reduced the expression of Bax, Bcl-2, cleaved caspase-3, IL-1ß, IL-6, and TNF-alpha. Our results showed that GSDMD knockdown can inhibit HG-induced mitochondrial ROS production and JNK phosphorylation. CONCLUSIONS This study indicates that GSDMD knockdown can attenuate HG-induced inflammation and apoptosis by inhibiting the phosphorylation of JNK via mitochondrial ROS.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Podocytes/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Caspase 3 , Cell Culture Techniques , Diabetic Nephropathies/metabolism , Glucose/metabolism , Inflammation , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mitochondria/metabolism , Podocytes/drug effects , Proto-Oncogene Proteins c-bcl-2 , Pyroptosis/drug effects , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein
13.
JCI Insight ; 52019 03 07.
Article in English | MEDLINE | ID: mdl-30843871

ABSTRACT

The endoplasmic reticulum (ER) of cancer cells needs to adapt to the enhanced proteotoxic stress associated with the accumulation of unfolded, misfolded and transformation-associated proteins. One way by which tumors thrive in the context of ER stress is by promoting ER-Associated Degradation (ERAD), although the mechanisms are poorly understood. Here, we show that the Small p97/VCP Interacting Protein (SVIP), an endogenous inhibitor of ERAD, undergoes DNA hypermethylation-associated silencing in tumorigenesis to achieve this goal. SVIP exhibits tumor suppressor features and its recovery is associated with increased ER stress and growth inhibition. Proteomic and metabolomic analyses show that cancer cells with epigenetic loss of SVIP are depleted in mitochondrial enzymes and oxidative respiration activity. This phenotype is reverted upon SVIP restoration. The dependence of SVIP hypermethylated cancer cells on aerobic glycolysis and glucose was also associated with sensitivity to an inhibitor of the glucose transporter GLUT1. This could be relevant to the management of tumors carrying SVIP epigenetic loss, because these occur in high-risk patients who manifest poor clinical outcomes. Overall, our study provides insights into how epigenetics helps deal with ER stress and how SVIP epigenetic loss in cancer may be amenable to therapies that target glucose transporters.


Subject(s)
Cellular Reprogramming/physiology , Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Epigenomics , Membrane Proteins/metabolism , Neoplasms/metabolism , Phosphate-Binding Proteins/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , Cell Survival/drug effects , Cellular Reprogramming/genetics , DNA Methylation , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic , Gene Silencing , Glucose Transporter Type 1 , Humans , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasms/genetics , Phenotype , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/pharmacology , Proteomics
15.
J Cell Physiol ; 229(2): 153-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23955241

ABSTRACT

Although neurons are not productively infected with HIV-1, neuronal injury and death are frequently seen in the brains of AIDS patients with neurological and neurocognitive disorders. Evidently, viral proteins including Tat and cellular inflammatory factors released by activated and/or infected microglia, macrophages, and astrocytes contribute to neuronal cell death. Several studies have demonstrated that HIV-1 associated neuronal cell injury is mediated by dysregulation of signaling pathways that are controlled, in part, by a class of serine/threonine kinases. In this study, we demonstrate that pDING, a novel plant-derived phosphate binding protein has the capacity to reduce the severity of injury and death caused by HIV-1 and its neurotoxic Tat protein. We demonstrate that pDING, also called p27SJ/p38SJ, protects cells from the loss of neuronal processes induced by Tat and promotes neuronal outgrowth after Tat-mediated injury. Further, expression of pDING prevents Tat-induced oxidative stress and mitochondrial permeability. With its profound phosphatase activity, pDING controls the activity of several kinases including MAPK, Cdk5, and their downstream target protein, MEF2, which is implicated in neuronal cell protection. Our results show that expression of pDING in neuronal cells diminishes the level of hyperphosphorylated forms of Cdk5 and MEF2 caused by Tat and the other neurotoxic agents that are secreted by the HIV-1 infected cells. These observations suggest that pDING, through its phosphatase activity, has the ability to manipulate the state of phosphorylation and activity of several factors involved in neuronal cell health in response to HIV-1.


Subject(s)
Neurons/drug effects , Phosphate-Binding Proteins/pharmacology , Plant Proteins/pharmacology , tat Gene Products, Human Immunodeficiency Virus/toxicity , Cell Death , Cells, Cultured , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Humans , Hypericum/chemistry , Mitochondria/drug effects , Mitochondria/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Oxidative Stress/drug effects , Phosphate-Binding Proteins/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism
16.
J Ren Nutr ; 18(6): 521-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18940656

ABSTRACT

OBJECTIVE: To evaluate the effectiveness of a protocol designed to optimize serum phosphate levels in patients undergoing regular hemodialysis (HD). DESIGN: Randomized, controlled trial. SETTING: Hemodialysis units at Barts and the London NHS Trust and satellite units. PATIENTS: Thirty-four clinically stable adults undergoing regular HD with a serum phosphate level >1.8 mmol/L on at least one occasion within 4 months of starting the study. INTERVENTION: Management of serum phosphate using a specially designed phosphate management protocol during a 4-month study period implemented by a renal dietitian and renal pharmacist compared with standard practice. MAIN OUTCOME MEASURE: Change in serum phosphate levels in both groups after 4 months. RESULTS: Patients managed using the phosphate management protocol had a significantly greater reduction in serum phosphate levels compared with patients receiving standard practice (-0.22 +/- 0.67 mmol/L vs. +0.19 +/- 0.32 mmol/L, P = 0.03). CONCLUSION: The phosphate management protocol was effective, and its implementation was associated with significantly better serum phosphate control in patients undergoing regular HD.


Subject(s)
Kidney Failure, Chronic/blood , Kidney Failure, Chronic/therapy , Phosphate-Binding Proteins/pharmacology , Phosphates/blood , Renal Dialysis , Calcium/blood , Calcium/metabolism , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Parathyroid Hormone/metabolism , Patient Education as Topic , Patient Participation , Renal Dialysis/adverse effects , Treatment Outcome , Vitamin D/blood , Vitamin D/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...