Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 495
Filter
1.
Respir Res ; 25(1): 355, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354535

ABSTRACT

INTRODUCTION: Pyroptosis, inflammatory necrosis of cells, is a programmed cell death involved in the pathological process of diseases. Endoplasmic reticulum stress (ERS), as a protective stress response of cell, decreases the unfold protein concentration to inhibit the unfold protein agglutination. Whereas the relationship between endoplasmic reticulum stress and pyroptosis in pulmonary hypertension (PH) remain unknown. Previous evident indicated that circular RNA (circRNA) can participate in several biological process, including cell pyroptosis. However, the mechanism of circRNA regulate pyroptosis of pulmonary artery smooth muscle cells through endoplasmic reticulum stress still unclear. Here, we proved that circSSR1 was down-regulate expression during hypoxia in pulmonary artery smooth muscle cells, and over-expression of circSSR1 inhibit pyroptosis both in vitro and in vivo under hypoxic. Our experiments have indicated that circSSR1 could promote host gene SSR1 translation via m6A to activate ERS leading to pulmonary artery smooth muscle cell pyroptosis. In addition, our results showed that G3BP1 as upstream regulator mediate the expression of circSSR1 under hypoxia. These results highlight a new regulatory mechanism for pyroptosis and provide a potential therapy target for pulmonary hypertension. METHODS: RNA-FISH and qRT-PCR were showed the location of circSSR1 and expression change. RNA pull-down and RIP verify the circSSR1 combine with YTHDF1. Western blotting, PI staining and LDH release were used to explore the role of circSSR1 in PASMCs pyroptosis. RESULTS: CircSSR1 was markedly downregulated in hypoxic PASMCs. Knockdown CircSSR1 inhibited hypoxia induced PASMCs pyroptosis in vivo and in vitro. Mechanistically, circSSR1 combine with YTHDF1 to promote SSR1 protein translation rely on m6A, activating pyroptosis via endoplasmic reticulum stress. Furthermore, G3BP1 induce circSSR1 degradation under hypoxic. CONCLUSION: Our findings clarify the role of circSSR1 up-regulated parental protein SSR1 expression mediate endoplasmic reticulum stress leading to pyroptosis in PASMCs, ultimately promoting the development of pulmonary hypertension.


Subject(s)
Endoplasmic Reticulum Stress , Myocytes, Smooth Muscle , Pulmonary Artery , Pyroptosis , Endoplasmic Reticulum Stress/physiology , Pyroptosis/physiology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Animals , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , RNA, Circular/metabolism , RNA, Circular/genetics , Male , Cells, Cultured , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/genetics , Membrane Proteins
2.
FASEB J ; 38(18): e70054, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39297783

ABSTRACT

Excessive proinflammatory cytokine release induced by pyroptosis plays a vital role in intestinal mucosal inflammation in ulcerative colitis (UC). Several pyroptosis-related factors are regulated by the centrosome. Pericentriolar material 1 (PCM1) is a primary component of centriolar satellites that is present as cytoplasmic granules around the centrosome. Our previous study revealed that PCM1 was highly expressed in UC patients, but the role of PCM1 in UC remains unknown. This study aimed to elucidate the role of PCM1 in the development of UC, especially the mechanism in pyroptosis process of UC. Clinical mucosal sample and dextran sulfate sodium (DSS)-induced colitis mouse were used to reveal the association between PCM1 and intestinal inflammation. Intestinal epithelial cell-specific PCM1-knockout mice were constructed to determine the role of PCM1 in colitis. Finally, PCM1 RNA interference and overexpression assays in THP1 cells were employed to study the molecular mechanisms of PCM1 in inflammatory responses and pyroptosis. We found that PCM1 expression was upregulated in the colonic mucosa of UC patients and positively correlated with inflammatory indicators. PCM1 expression was elevated in DSS-induced colitis mice and was reduced after methylprednisolone treatment. In the DSS colitis model, intestinal-specific PCM1-knockout mice exhibited milder intestinal inflammation and lower pyroptosis levels than wild-type mice. In cell level, PCM1 exerted a proinflammatory effect by activating the NLRP3 inflammasome and triggering subsequent gasdermin D-mediated pyroptosis to release IL-1ß and IL-18. In conclusion, PCM1 mediates activation of the NLRP3 inflammasome and gasdermin D-dependent pyroptosis, ultimately accelerating intestinal inflammation in UC. These findings revealed a previously unknown role of PCM1 in initiating intestinal mucosal inflammation and pyroptosis in UC, and this factor is expected to be a regulator in the complex inflammatory network of UC.


Subject(s)
Colitis, Ulcerative , Intracellular Signaling Peptides and Proteins , Macrophages , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphate-Binding Proteins , Pyroptosis , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pyroptosis/physiology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Mice , Humans , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/metabolism , Male , Mice, Inbred C57BL , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Female , Dextran Sulfate/toxicity , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Gasdermins
3.
J Obstet Gynaecol Res ; 50(10): 1813-1829, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39288911

ABSTRACT

INTRODUCTION: Hyperglycemia is closely related to trophoblast dysfunction during pregnancy and results in suppressed invasion, migration, and pro-inflammatory cell death of trophoblasts. Hyperglycemia is a dependent risk factor for gestational hypertension accompanied by decreased placental growth factor (PLGF), which is important for maternal and fetal development. However, there is currently a lack of evidence to support whether PLGF can alleviate trophoblast cell dysfunction caused by high blood sugar. Here, we aim to clarify the effect of hyperglycemia on trophoblast dysfunction and determine how PLGF affects this process. METHODS: The changes in placental tissue histomorphology from gestational diabetes mellitus (GDM) patients were compared with those of normal placentas. HTR8/SVneo cells were cultured in different amounts of glucose to examine cellular pyroptosis, migration, and invasion as well as PLGF levels. Furthermore, the levels of pyroptosis-related proteins (NLRP3, pro-caspase1, caspase1, IL-1ß, and Gasdermin D [GSDMD]) as well as autophagy-related proteins (LC3-II, Beclin1, and p62) were examined by Western blotting. The GFP-mRFP-LC3-II system and transmission electron microscopy were used to detect mitophagy levels, and small interfering RNAs targeting BCL2 Interacting Protein 3 (siBNIP3) and PTEN-induced kinase 1 (siPINK1) were used to determine the role of mitophagy in pyroptotic death of HTR-8/SVneo cells. RESULTS: Our results show that hyperglycemia upregulates NLRP3, pro-caspase1, caspase1, IL-1ß at the protein level in GDM patients. High glucose (HG, 25 mM) inhibits viability, invasion, and migration of trophoblast cells while suppressing superoxide dismutase levels and promoting malondialdehyde production, thus leading to a senescence associated beta-gal-positive cell burst. PLGF levels in nucleus and the cytosol are also inhibited by HG, whereas PLGF treatment inhibited pyroptosis-related protein levels of NLRP3, pro-caspase1, caspase1, IL-1ß, and GSDMD, Gasdermin D N-terminal domain (GSDMD-N). HG-induced mitochondrial dysfunction and BNIP3 and PINK1/Parkin expression. Knocking down BINP3 and PINK1 abolished the protective role of PLGF by preventing mitophagy. CONCLUSION: PLGF inhibited hyperglycemia, while PLGF reversed hyperglycemic injury by promoting mitophagy via the BNIP3/PINK1/Parkin pathway. Altogether, these results suggest that PLGF may protect against trophoblast dysfunction in diabetes.


Subject(s)
Diabetes, Gestational , Hyperglycemia , Mitophagy , Placenta Growth Factor , Pyroptosis , Trophoblasts , Humans , Pyroptosis/drug effects , Pyroptosis/physiology , Trophoblasts/metabolism , Female , Pregnancy , Placenta Growth Factor/metabolism , Diabetes, Gestational/metabolism , Hyperglycemia/metabolism , Mitophagy/drug effects , Adult , Cell Line
4.
Medicina (Kaunas) ; 60(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39336475

ABSTRACT

Background and Objectives: Fine particulate matter, PM2.5, is becoming a major threat to human health, particularly in terms of respiratory diseases. Pyroptosis is a recently discovered and distinct form of cell death, characterized by pore formation in the cell membrane and secretions of proinflammatory cytokines. There has been little research on the effect of PM2.5 on pyroptosis, especially in airway epithelium. We investigated whether PM2.5-related oxidative stress induces pyroptosis in bronchial epithelial cells and defined the underlying mechanisms. Materials and Methods: After exposure of a BEAS-2B cell line to PM2.5 concentration of 20 µg/mL, reactive oxygen species (ROS) levels, parameters related to pyroptosis, and NF-κB signaling were measured by Western blotting, immunofluorescence, and ELISA (Enzyme-linked immunosorbent assay). Results: PM2.5 induced pyroptotic cell death, accompanied by LDH (Lactate dehydrogenase) release and increased uptake of propidium iodide in a dose-dependent manner. PM2.5 activated the NLRP3-casp1-gasdermin D pathway, with resulting secretions of the proinflammatory cytokines IL-1ß and IL-18. The pyroptosis activated by PM2.5 was alleviated significantly by NLRP3 inhibitor. In PM2.5-exposed BEAS-2B cells, levels of intracellular ROS and NF-κB p65 increased. ROS scavenger inhibited the expression of the NLRP3 inflammasome, and the NF-κB inhibitor attenuated pyroptotic cell death triggered by PM2.5 exposure, indicating that the ROS/NF-κB pathway is involved in PM2.5-induced pyroptosis. Conclusions: These findings show that PM2.5 exposure can cause cell injury by NLRP3-inflammasome-mediated pyroptosis by upregulating the ROS/NF-κB pathway in airway epithelium.


Subject(s)
Epithelial Cells , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Particulate Matter , Pyroptosis , Reactive Oxygen Species , Signal Transduction , Pyroptosis/drug effects , Pyroptosis/physiology , Humans , Particulate Matter/adverse effects , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Cell Line , Bronchi/drug effects , Bronchi/metabolism , Oxidative Stress/drug effects , Interleukin-1beta/metabolism , Interleukin-18/metabolism
5.
Brain Res Bull ; 217: 111060, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39236791

ABSTRACT

BACKGROUND: The molecular biology mechanisms underlying postoperative cognitive dysfunction (POCD) remain unclear, resulting in a lack of specific therapeutic targets and limited clinical treatment options. The NLRP3 pyroptotic pathway, induced by neuroinflammation, is known to promote the development of POCD. Research has shown that lncRNA MEG3 exacerbates cell pyroptosis in various neurological injuries, though the precise mechanism remains to be investigated. METHODS: In vitro and in vivo models of POCD were established through treatment with sevoflurane. Gene and protein expression were investigated using qRT-PCR, Western blot analysis, ELISA, and histological staining. Additionally, cell viability and injury were assessed through CCK-8 and LDH assays. Hippocampal-dependent memory and cognitive abilities were evaluated using the Morris Water Maze (MWM) test. Furthermore, the interactions between MEG3 and EZH2/YTHDC1 were validated through RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP). RESULTS: Our findings reveal that sevoflurane significantly reduced MEG3 and pyroptosis-related proteins in mice. The overexpression of MEG3 protected mice against sevoflurane-induced cognitive dysfunction and reversed sevoflurane-induced pyroptosis in hippocampal neurons. MEG3 induced the downregulation of NLRP3 expression and reduced mRNA stability through its interaction with EZH2/YTHDC1. CONCLUSION: In conclusion, our study elucidates that MEG3 inhibits the NLRP3 inflammasome and hippocampal neuron pyroptosis through the recruitment of EZH2/YTHDC1. These findings shed light on the underlying mechanism of MEG3 in the regulation of POCD and suggest that MEG3 could serve as a potential therapeutic target for the treatment of POCD.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Hippocampus , Postoperative Cognitive Complications , Pyroptosis , RNA, Long Noncoding , Sevoflurane , Animals , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Pyroptosis/physiology , Mice , Postoperative Cognitive Complications/metabolism , Hippocampus/metabolism , Male , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Sevoflurane/pharmacology , Mice, Inbred C57BL , Neurons/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Cognitive Dysfunction/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
6.
J Orthop Surg Res ; 19(1): 550, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252111

ABSTRACT

Osteoarthritis (OA) is a chronic joint disease characterized by the degeneration, destruction, and excessive ossification of articular cartilage. The prevalence of OA is rising annually, concomitant with the aging global population and increasing rates of obesity. This condition imposes a substantial and escalating burden on individual health, healthcare systems, and broader social and economic frameworks. The etiology of OA is multifaceted and not fully understood. Current research suggests that the death of chondrocytes, encompassing mechanisms such as cellular apoptosis, pyroptosis, autophagy, ferroptosis and cuproptosis, contributes to both the initiation and progression of the disease. These cell death pathways not only diminish the population of chondrocytes but also exacerbate joint damage through the induction of inflammation and other deleterious processes. This paper delineates the morphological characteristics associated with various modes of cell death and summarizes current research results on the molecular mechanisms of different cell death patterns in OA. The objective is to review the advancements in understanding chondrocyte cell death in OA, thereby offering novel insights for potential clinical interventions.


Subject(s)
Cell Death , Chondrocytes , Disease Progression , Osteoarthritis , Chondrocytes/pathology , Humans , Osteoarthritis/pathology , Osteoarthritis/therapy , Cell Death/physiology , Apoptosis/physiology , Cartilage, Articular/pathology , Autophagy/physiology , Animals , Pyroptosis/physiology , Ferroptosis/physiology
7.
ASN Neuro ; 16(1): 2394352, 2024.
Article in English | MEDLINE | ID: mdl-39249102

ABSTRACT

Iron is a critical transition metal required to sustain a healthy central nervous system. Iron is involved in metabolic reactions, enzymatic activity, myelinogenesis, and oxygen transport. However, in several pathological conditions such as cancer, neurodegeneration, and neurotrauma iron becomes elevated. Excessive iron can have deleterious effects leading to reactive oxygen species (ROS) via the Fenton reaction. Iron-derived ROS are known to drive several mechanisms such as cell death pathways including ferroptosis, necroptosis, and pyroptosis. Excessive iron present in the post-traumatic brain could trigger these harmful pathways potentiating the high rates of morbidity and mortality. In the present review, we will discuss how iron plays an intricate role in initiating ferroptosis, necroptosis, and pyroptosis, examine their potential link to traumatic brain injury morbidity and mortality, and suggest therapeutic targets.


Subject(s)
Brain Injuries, Traumatic , Ferroptosis , Iron , Necroptosis , Pyroptosis , Pyroptosis/physiology , Humans , Ferroptosis/physiology , Iron/metabolism , Necroptosis/physiology , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Reactive Oxygen Species/metabolism
8.
Biomed Pharmacother ; 179: 117344, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39191021

ABSTRACT

Osteoarthritis (OA) is the most common degenerative joint disease. Multiple tissues are altered during the development of OA, resulting in joint pain and permanent damage to the osteoarticular joints. Current research has demonstrated that non-apoptotic cell death plays a crucial role in OA. With the continuous development of targeted therapies, non-apoptotic cell death has shown great potential in the prevention and treatment of OA. We systematically reviewed research progress on the role of non-apoptotic cell death in the pathogenesis, development, and outcome of OA, including autophagy, pyroptosis, ferroptosis, necroptosis, immunogenic cell death, and parthanatos. This article reviews the mechanism of non-apoptotic cell death in OA and provides a theoretical basis for the identification of new targets for OA treatment.


Subject(s)
Autophagy , Cell Death , Osteoarthritis , Osteoarthritis/pathology , Humans , Animals , Cell Death/physiology , Autophagy/physiology , Pyroptosis/physiology , Ferroptosis/physiology , Necroptosis , Apoptosis
9.
Biomed Pharmacother ; 179: 117367, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39214011

ABSTRACT

The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.


Subject(s)
Heart Failure , Myocytes, Cardiac , NF-kappa B , Pyroptosis , Signal Transduction , Pyroptosis/physiology , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Heart Failure/metabolism , Heart Failure/pathology , Animals
10.
Neurochem Res ; 49(11): 3105-3117, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39167346

ABSTRACT

Cerebral ischemia reperfusion injury is a severe neurological impairment that occurs after blood flow reconstruction in stroke, and microglia cell pyroptosis is one of its important mechanisms. Electroacupuncture has been shown to be effective in mitigating and alleviating cerebral ischemia reperfusion injury by inhibiting neuroinflammation, reducing cellular pyroptosis, and improving neurological function. In this experiment, we divided the rats into three groups, including the sham operation (Sham) group, the middle cerebral artery occlusion/reperfusion (MCAO/R) group, and the pre-electroacupuncture (EAC) group. Pre-electroacupuncture group was stimulated with electroacupuncture of a certain intensity on the Baihui (GV 20) and Dazhui (GV 14) of the rat once a day from the 7th day to the 1st day before the MCAO/R operation. The extent of cerebral infarction was detected by TTC staining. A modified Zea-Longa five-point scale scoring system was used to determine neurologic function in MCAO rats. The number of neurons and morphological changes were accessed by Nissl staining and HE staining. The cellular damage was detected by TUNEL staining. In addition, the expression levels of RhoA, pyrin, GSDMD, Caspase1, cleaved-Caspase1, Iba-1, CD206, and ROCK2 were examined by western blotting and immunofluorescence. The results found that pre-electroacupuncture significantly attenuated neurological impairment and cerebral infarction compared to the post-MCAO/R rats. In addition, pre-electroacupuncture therapy promoted polarization of microglia to the neuroprotective (M2) phenotype. In addition, pre-electroacupuncture inhibited microglia pyroptosis by inhibiting RhoA/pyrin/GSDMD signaling pathway, thereby reducing neuronal injury and increasing neuronal survival in the MCAO/R rats. Taken together, these results demonstrated that pre-acupuncture could attenuate cerebral ischemia-reperfusion injury by inhibiting microglial pyroptosis. Therefore, pre-electroacupuncture might be a potential preventive strategy for ischemic stroke patients.


Subject(s)
Electroacupuncture , Microglia , Reperfusion Injury , Signal Transduction , Animals , Male , Rats , Brain Ischemia/metabolism , Brain Ischemia/therapy , Electroacupuncture/methods , Gasdermins , Infarction, Middle Cerebral Artery/therapy , Infarction, Middle Cerebral Artery/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphate-Binding Proteins/metabolism , Pyroptosis/physiology , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/therapy , Reperfusion Injury/prevention & control , rho GTP-Binding Proteins , rhoA GTP-Binding Protein/metabolism , Signal Transduction/physiology
11.
Pathol Res Pract ; 261: 155490, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39126977

ABSTRACT

Pyroptosis is an inflammatory programed cell death process that plays a crucial role in cancer therapeutic, while Gasdermin-D is a critical effector protein for pyroptosis execution. This review discusses the intricate interactions between Gasdermin-D and some non-coding RNAs (lncRNA, miRNA, siRNA) and their potential application in the regulation of pyroptosis as an anticancer therapy. Correspondingly, these ncRNAs significantly implicate in Gasdermin-D expression and function regarding the pyroptosis pathway. Functioning as competing endogenous RNAs (ceRNAs), these ncRNAs might regulate Gasdermin-D at the molecular level, underlying fatal cell death caused by cancer and tumor propagation. Therefore, these interactions appeal to therapeutics, offering new avenues for cancer treatment. It address this research gap by discussing the possible roles of ncRNAs as mediators of gasdermin-D regulation. It suggest therapeutic strategies based on the current research findings to ensure the interchange between the ideal pyroptosis and cancer cell death.


Subject(s)
Intracellular Signaling Peptides and Proteins , Neoplasms , Phosphate-Binding Proteins , Pyroptosis , RNA, Untranslated , Pyroptosis/physiology , Humans , Neoplasms/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Animals , Gene Expression Regulation, Neoplastic , Gasdermins
12.
Exp Eye Res ; 246: 110019, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39117137

ABSTRACT

Cataracts are the world's number one blinding eye disease. Cataracts can only be effectively treated surgically, although there is a chance of surgical complications. One of the pathogenic processes of cataracts is oxidative stress, which closely correlated with pyroptosis. SIRT1 is essential for the regulation of pyroptosis. Nevertheless, the role of SIRT1 in formation of cataracts is unclear. In this work, we developed an in vitro model of shortwave blue light (SWBL)-induced scotomization in human lens epithelial cells (HLECs) and an in vivo model of SWBL-induced cataracts in rats. The study aimed to understand how the SIRT1/NF-κB/NLRP3 pathway functions. Additionally, the evaluation included cell death and the release of lactate dehydrogenase (LDH), a cytotoxicity marker, from injured cells. First, we discovered that SWBL exposure resulted in lens clouding in Sprague- Dawley (SD) rats and that the degree of clouding was positively linked to the duration of irradiation. Second, we discovered that SIRT1 exhibited antioxidant properties and was connected to the NF-κB/NLRP3 pathway. SWBL irradiation inhibited SIRT1 expression, exacerbated oxidative stress, and promoted nuclear translocation of NF-κB and the activation of the NLRP3 inflammasome, which caused LEC pyroptosis and ultimately led to cataract formation. Transient transfection to increase the expression of SIRT1 decreased the protein expression levels of NF-κB, NLRP3, caspase-1, and GSDMD, inhibited HLEC pyroptosis, and reduced the release of LDH, providing a potential method for cataract prevention and treatment.


Subject(s)
Cataract , Epithelial Cells , Lens, Crystalline , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Sirtuin 1 , Animals , Humans , Rats , Blotting, Western , Blue Light/adverse effects , Cataract/metabolism , Cataract/pathology , Cataract/etiology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Lens, Crystalline/radiation effects , Lens, Crystalline/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Pyroptosis/physiology , Pyroptosis/radiation effects , Rats, Sprague-Dawley , Signal Transduction/physiology , Sirtuin 1/metabolism
14.
J Diabetes Complications ; 38(10): 108848, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39178624

ABSTRACT

Diabetes is a major risk factor for cardiovascular diseases, and myocardial damage caused by hyperglycemia is the main cause of heart failure. However, there is still a lack of systematic understanding of myocardial damage caused by diabetes. At present, we believe that the cellular inflammatory damage caused by hyperglycemia is one of the causes of diabetic cardiomyopathy. Pyroptosis, as a proinflammatory form of cell death, is closely related to the occurrence and development of diabetic cardiomyopathy. Therefore, this paper focuses on the important role of inflammation in the occurrence and development of diabetic cardiomyopathy. From the perspective of pyroptosis, we summarize the pyroptosis of different types of cells in diabetic cardiomyopathy and its related signaling pathways. It also summarizes the treatment of diabetic cardiomyopathy, hoping to provide methods for the prevention and treatment of diabetic cardiomyopathy by inhibiting pyroptosis.


Subject(s)
Diabetic Cardiomyopathies , Pyroptosis , Pyroptosis/drug effects , Pyroptosis/physiology , Diabetic Cardiomyopathies/therapy , Diabetic Cardiomyopathies/prevention & control , Humans , Animals , Signal Transduction/drug effects , Myocardium/pathology , Myocardium/metabolism
15.
Arch Pharm Res ; 47(7): 617-631, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987410

ABSTRACT

Regulated cell death (RCD) pathways, such as pyroptosis, apoptosis, and necroptosis, are essential for maintaining the body's balance, defending against pathogens, and eliminating abnormal cells that could lead to diseases like cancer. Although these pathways operate through distinct mechanisms, recent genetic and pharmacological studies have shown that they can interact and influence each other. The concept of "PANoptosis" has emerged, highlighting the interplay between pyroptosis, apoptosis, and necroptosis, especially during cellular responses to infections. This article provides a concise overview of PANoptosis and its molecular mechanisms, exploring its implications in various diseases. The review focuses on the extensive interactions among different RCD pathways, emphasizing the role of PANoptosis in infections, cytokine storms, inflammatory diseases, and cancer. Understanding PANoptosis is crucial for developing novel treatments for conditions involving infections, sterile inflammations, and cancer.


Subject(s)
Inflammation , Necroptosis , Neoplasms , Pyroptosis , Humans , Inflammation/pathology , Inflammation/drug therapy , Inflammation/immunology , Animals , Necroptosis/drug effects , Necroptosis/physiology , Pyroptosis/drug effects , Pyroptosis/physiology , Neoplasms/pathology , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Apoptosis/drug effects , Cell Death/physiology , Regulated Cell Death/drug effects , Infections/pathology , Infections/immunology
16.
J Neuroimmunol ; 393: 578401, 2024 08 15.
Article in English | MEDLINE | ID: mdl-38996718

ABSTRACT

BACKGROUND: We previously reported that the HMGB1/TLR4 axis promoted inflammation during the acute phase of intracerebral hemorrhage. Given that this phase is known to involve neuronal pyroptosis and neuroinflammation, here we explore whether HMGB1/TLR signaling activate inflammasome and pyroptosis after intracerebral hemorrhage. METHODS: Autologous blood was injected into Sprague-Dawley rats to induce intracerebral hemorrhage. Neurological deficits were assessed using a modified neurological severity score. These expression and localization of NLRP1 and NLRP3 inflammasomes, as well as the levels of pyroptosis and pyroptosis-associated proteins were assessed using Western blot or immunocytochemistry. These experiments were repeated in animals that received treatment with short interfering RNAs against NLRP1 or NLRP3, with HMGB1 inhibitor ethyl pyruvate or TLR4 inhibitor TAK-242. RESULTS: Intracerebral hemorrhage upregulated NLRP1 and NLRP3 in the ipsilateral striatum and increased the proportions of these cells that were pyroptosis-positive. Additionally, the levels of caspase protein family (e.g., pro-caspase-1 and caspase-1), apoptosis-associated speck-like protein (ASC), pro-interleukin-1ß (IL-1ß), and IL-1ß were also elevated. These effects on pyroptosis and associated neurological deficit, were partially reversed by knockdown of NLRP1 or NLRP3, or by inhibition of HMGB1 or TLR4. Inhibition of HMGB1 or TLR4 resulted in the downregulation NLRP3 but not NLRP1. CONCLUSIONS: The HMGB1/TLR4 signaling may activate the NLRP3 inflammasome during the acute phase of intracerebral hemorrhage, resulting in the inflammatory process known as pyroptosis. These insights suggest potential therapeutic targets for the mitigation tissue injury and associated neurological deficits following hemorrhagic stroke.


Subject(s)
Cerebral Hemorrhage , HMGB1 Protein , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Rats, Sprague-Dawley , Toll-Like Receptor 4 , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/physiology , Pyroptosis/drug effects , HMGB1 Protein/metabolism , Rats , Toll-Like Receptor 4/metabolism , Male , Cerebral Hemorrhage/metabolism , Inflammasomes/metabolism , Signal Transduction/physiology , Signal Transduction/drug effects , Nerve Tissue Proteins
17.
Exp Neurol ; 380: 114900, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39059736

ABSTRACT

AIMS: Adipose-derived stem cell (ADSC)-derived exosomes have been recognized for their neuroprotective effects in various neurological diseases. This study investigates the potential neuroprotective effects of ADSC-derived exosomes in sepsis-associated encephalopathy (SAE). METHODS: Behavioral cognitive functions were evaluated using the open field test, Y-maze test, and novel object recognition test. Brain activity was assessed through functional magnetic resonance imaging (fMRI). Pyroptosis was measured using immunofluorescence staining and western blotting. RESULTS: Our findings indicate that ADSC-derived exosomes mitigate cognitive impairment, improve survival rates, and prevent weight loss in SAE mice. Additionally, exosomes protect hippocampal function in SAE mice, as demonstrated by fMRI evaluations. Furthermore, SAE mice exhibit neuronal damage and infiltration of inflammatory cells in the hippocampus, conditions which are reversed by exosome treatment. Moreover, our study highlights the downstream regulatory role of the NLRP3/caspase-1/GSDMD signaling pathway as a crucial mechanism in alleviating hippocampal inflammation. CONCLUSION: ADSC-derived exosomes confer neuroprotection in SAE models by mediating the NLRP3/caspase-1/GSDMD pathway, thereby ameliorating cognitive impairment.


Subject(s)
Caspase 1 , Exosomes , Hippocampus , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Sepsis-Associated Encephalopathy , Animals , Pyroptosis/physiology , Exosomes/metabolism , Exosomes/transplantation , Hippocampus/metabolism , Hippocampus/pathology , Sepsis-Associated Encephalopathy/metabolism , Mice , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase 1/metabolism , Neuroprotection/physiology , Gasdermins , Phosphate-Binding Proteins
18.
Respir Res ; 25(1): 291, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080660

ABSTRACT

Acute lung injury (ALI) is characterized by an unregulated inflammatory reaction, often leading to severe morbidity and ultimately death. Excessive inflammation caused by M1 macrophage polarization and pyroptosis has been revealed to have a critical role in ALI. Recent study suggests that glycolytic reprogramming is important in the regulation of macrophage polarization and pyroptosis. However, the particular processes underlying ALI have yet to be identified. In this study, we established a Lipopolysaccharide(LPS)-induced ALI model and demonstrated that blocking glycolysis by using 2-Deoxy-D-glucose(2-DG) significantly downregulated the expression of M1 macrophage markers and pyroptosis-related genes, which was consistent with the in vitro results. Furthermore, our research has revealed that Phosphoglycerate Kinase 1(PGK1), an essential enzyme in the glycolysis pathway, interacts with NOD-, LRR- and pyrin domain-containing protein 3(NLRP3). We discovered that LPS stimulation improves the combination of PGK1 and NLRP3 both in vivo and in vitro. Interestingly, the absence of PGK1 reduces the phosphorylation level of NLRP3. Based on in vitro studies with mice bone marrow-derived macrophages (BMDMs), we further confirmed that siPGK1 plays a protective role by inhibiting macrophage pyroptosis and M1 macrophage polarization. The PGK1 inhibitor NG52 suppresses the occurrence of excessive inflammation in ALI. In general, it is plausible to consider a therapeutic strategy that focuses on modulating the relationship between PGK1 and NLRP3 as a means to mitigate the activation of inflammatory macrophages in ALI.


Subject(s)
Acute Lung Injury , Macrophages , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphoglycerate Kinase , Pyroptosis , Pyroptosis/physiology , Pyroptosis/drug effects , Animals , Phosphoglycerate Kinase/metabolism , Phosphoglycerate Kinase/genetics , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Mice , Macrophages/metabolism , Macrophages/drug effects , Macrophages/enzymology , Glycolysis/physiology , Glycolysis/drug effects , Male , Lipopolysaccharides/toxicity , Mice, Knockout , Cells, Cultured
19.
J Sex Med ; 21(8): 652-662, 2024 08 01.
Article in English | MEDLINE | ID: mdl-38972660

ABSTRACT

BACKGROUND: Diabetes mellitus commonly causes endothelial cell and smooth muscle cell death in penile cavernous tissue. AIM: The study sought to study the mode of cell death in the penile cavernous tissue in type 1 diabetic rats. METHODS: A total of 36 Sprague Dawley rats 10 weeks of age were randomly divided into 2 groups: a normoglycemic group and type 1 diabetic group (intraperitoneal injection of Streptozotocin (STZ), 60 mg/kg). We randomly selected 6 rats from each group for tests at the end of 11, 14, and 18 weeks of age, respectively. All rats were able to eat and drink freely. The ratio of maximum intracavernous pressure to mean arterial pressure, concentration of serum testosterone, level of nitric oxide in the penile cavernosum, and expression of active caspase-1 (pyroptosis) and active caspase-3 (apoptosis) were determined. OUTCOMES: At the end of weeks 4 and 8 of type 1 diabetes, the proportions of endothelial cells and smooth muscle cells undergoing apoptosis and pyroptosis in penile cavernous tissue are different. RESULTS: The ratio of maximum intracavernous pressure to mean arterial pressure and nitric oxide levels were significantly lower in the 4- and 8-week diabetic groups than in the normoglycemic group (P < .01). Penile endothelial cell pyroptosis (5.67 ± 0.81%), smooth muscle cell apoptosis (23.72 ± 0.48%), total cell pyroptosis (9.67 ± 0.73%), and total apoptosis (10.52 ± 1.45%) were significantly greater in the 4-week diabetic group than in the normoglycemic group (P < .01). The proportion of endothelial cell pyroptosis (24.4 ± 3.69%), endothelial cell apoptosis (22.13 ± 2.43%), total cell pyroptosis (14.75 ± 0.93%), and total apoptosis (14.82 ± 1.08%) in the penile tissues of the 8-week diabetic group were significantly greater than those in the normoglycemic group (P < .01).The 8-week survival proportions of diabetic endothelial cells (38.86 ± 8.85%) and smooth muscle cells (44.46 ± 2.94%) was significantly lower than the 4-week survival proportions of endothelial cells (93.17 ± 8.07%) and smooth muscle cells (75.12 ± 4.76%) (P < .05). CLINICAL TRANSLATION: Inhibition of cell death by different methods at different stages may be the key to the treatment of type 1 diabetes-induced erectile dysfunction. STRENGTHS AND LIMITATIONS: The effect of type 1 diabetes on other types of cell death in penile cavernous tissue needs further study. CONCLUSION: The mode of death of endothelial cells in the cavernous tissue of the penis in the early stage in diabetic rats is dominated by pyroptosis, and the death of smooth muscle cells is dominated by apoptosis. Endothelial cell and smooth muscle cell death are not consistent at different stages of diabetes progression.


Subject(s)
Apoptosis , Caspase 3 , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Nitric Oxide , Penis , Rats, Sprague-Dawley , Male , Animals , Penis/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/complications , Rats , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/pathology , Caspase 3/metabolism , Apoptosis/physiology , Nitric Oxide/metabolism , Pyroptosis/physiology , Testosterone/blood , Caspase 1/metabolism , Endothelial Cells/pathology , Cell Death
20.
Pathol Res Pract ; 260: 155444, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38986361

ABSTRACT

Lung cancer is still a global health challenge in terms of high incidence, morbidity, and mortality. Recent scientific studies have determined that pyroptosis, a highly inflammatory form of programmed cell death, can be identified as a potential lung cancer therapeutic target. The NLRP3 inflammasome acts as a critical mediator in this process and, upon activation, activates multiprotein complex formation as well as caspase-1 activation. This process, triggered by a release of pro-inflammatory cytokines, results in pyroptotic cell death. Also, the relationship between the NLRP3 inflammasome and lung cancer was justified by its influence on tumour growth or metastasis. The molecular pathways produce progenitive mediators and remake the tissue. Finally, targeting NLRP3 inflammasome for pyroptosis induction and inhibition of its activation appears to be a promising lung cancer treatment approach. This technique makes cancer treatment more promising and personalized. This review explores the role of NLRP3 inflammasome activation and its possibilities in lung cancer treatment.


Subject(s)
Inflammasomes , Lung Neoplasms , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/physiology , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Inflammasomes/metabolism , Animals
SELECTION OF CITATIONS
SEARCH DETAIL