1
|
Li S, Zhou X, Duan Q, Niu S, Li P, Feng Y, Zhang Y, Xu X, Gong SP, Cao H. Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. Int J Mol Sci 2025; 26:3252. [PMID: 40244103 PMCID: PMC11989900 DOI: 10.3390/ijms26073252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by lipid accumulation and immune cell infiltration, is linked to plaque formation and cardiovascular events. While traditionally associated with lipid metabolism and endothelial dysfunction, recent research highlights the roles of autophagy and clonal hematopoiesis (CH) in its pathogenesis. Autophagy, a cellular process crucial for degrading damaged components, regulates macrophage homeostasis and inflammation, both of which are pivotal in atherosclerosis. In macrophages, autophagy influences lipid metabolism, cytokine regulation, and oxidative stress, helping to prevent plaque instability. Defective autophagy exacerbates inflammation, impairs cholesterol efflux, and accelerates disease progression. Additionally, autophagic processes in endothelial cells and smooth muscle cells further contribute to atherosclerotic pathology. Recent studies also emphasize the interplay between autophagy and CH, wherein somatic mutations in genes like TET2, JAK2, and DNMT3A drive immune cell expansion and enhance inflammatory responses in atherosclerotic plaques. These mutations modify macrophage function, intensifying the inflammatory environment and accelerating atherosclerosis. Chaperone-mediated autophagy (CMA), a selective form of autophagy, also plays a critical role in regulating macrophage inflammation by degrading pro-inflammatory cytokines and oxidized low-density lipoprotein (ox-LDL). Impaired CMA activity leads to the accumulation of these substrates, activating the NLRP3 inflammasome and worsening inflammation. Preclinical studies suggest that pharmacologically activating CMA may mitigate atherosclerosis progression. In animal models, reduced CMA activity accelerates plaque instability and increases inflammation. This review highlights the importance of autophagic regulation in macrophages, focusing on its role in inflammation, plaque formation, and the contributions of CH. Building upon current advances, we propose a hypothesis in which autophagy, programmed cell death, and clonal hematopoiesis form a critical intrinsic axis that modulates the fundamental functions of macrophages, playing a complex role in the development of atherosclerosis. Understanding these mechanisms offers potential therapeutic strategies targeting autophagy and inflammation to reduce the burden of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shuanhu Li
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
| | - Xin Zhou
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shukun Niu
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Pengquan Li
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Yihan Feng
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Ye Zhang
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shou-Ping Gong
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Huiling Cao
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| |
Collapse
|
2
|
Qiu B, Zhang S, Ge S, Yu Z, Wang D, Li K, Yu X, Tang C, Du J, Jin H, Huang Y. Vascular smooth muscle cell-derived SO 2 sulphenylated interferon regulatory factor 1 to inhibit VSMC senescence. Front Pharmacol 2025; 16:1516885. [PMID: 40223932 PMCID: PMC11986361 DOI: 10.3389/fphar.2025.1516885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Background Vascular smooth muscle cell (VSMC) senescence is a critical driver of vascular aging and various age-related cardiovascular diseases. Endogenous sulfur dioxide (SO2), a newly identified key cardiovascular gaseous signaling mediator, accelerates collagen deposition and vascular remodeling in VSMCs when downregulated. However, its effects on VSMC senescence remain unclear. Objective This study focused on exploring the role of endogenous SO2 in VSMC senescence and its associated molecular pathways. Methods Aged mice (24 months old), VSMC-specific aspartate aminotransferase 1 (AAT1) knockout (VSMC-AAT1-KO) mice, D-galactose (D-gal)-treated aorta rings and rat VSMC line A7r5 were used in the experiments. AAT1 expression was detected by Western blot and single-cell RNA sequencing. Senescence markers Tp53, p21Cip/Waf, interleukin 1β (IL-1β) and IL6 expression were detected by Western blot and real-time quantitative PCR. Senescence-associated β-galactosidase (SA-β-gal) activity was detected using SA-β-gal staining kit. Sulphenylation of interferon regulatory factor 1 (IRF1) was detected using a biotin switch assay. The plasmid for mutant IRF1 (mutation of cysteine 83 to serine, C83S) were constructed by site-directed mutagenesis. Results The expression of AAT1, a key enzyme for SO2 production, was reduced in the aortic tissue of aged mice in comparison to young mice. VSMC-AAT1-KO mice exhibited elevated protein expression of senescence markers Tp53, p21Cip/Waf and γ-H2AX in the aortic tissue. AAT1 knockdown in VSMCs elevated expression of Tp53, p21Cip/Waf, IL-1β and IL-6, and enhanced SA-β-gal activity. While SO2 donor supplementation rescued VSMC senescence caused by AAT1 knockdown and blocked aortic ring aging induced by D-gal. Mechanistically, SO2 promoted IRF1 sulphenylation, inhibited IRF1 nuclear translocation, which in turn downregulated the expression of senescence markers and the activity of SA-β-gal. Furthermore, mutation of C83 in IRF1 abolished SO2-mediated IRF1 sulphenylation and blocked the inhibitory effect of SO2 on VSMC senescence. Conclusion Reduction of the endogenous SO2/AAT1 pathway played a crucial role in driving VSMC senescence. Endogenous SO2 counteracted VSMC senescence and vascular aging via the sulphenylation of IRF1 at C83.
Collapse
Affiliation(s)
- Bingquan Qiu
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, China
| | - Shangyue Zhang
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, China
| | - Shuang Ge
- Department of Blood Transfusion Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhengyu Yu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Deqing Wang
- Department of Blood Transfusion Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, China
| | - Xiaoqi Yu
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Health Science Center, Peking University, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, China
| |
Collapse
|
3
|
Shang X, Wei R, Yang D, Yu B, Zhang W. Bioinformatics identification and validation of pyroptosis-related gene for ischemic stroke. BMC Med Genomics 2025; 18:53. [PMID: 40091022 PMCID: PMC11912741 DOI: 10.1186/s12920-025-02119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Ischemic stroke (IS) is one of the common and frequent diseases with extremely high lethality and disability in the world, and there is no effective treatment at present. This study aimed to screen hub genes involved in cerebral ischemia/reperfusion injury (CIRI) and pyroptosis, and explore promising intervention targets. METHODS CIRI-related genes (GSE202659 and GSE131193) and pyroptosis-related genes (PRGs) in mice were obtained from the Gene Expression Omnibus (GEO) and GeneCards database. We screened for LASSO regression to construct a prognostic model of GSE131193 and PRGs and examined by GSE137482. The functional enrichment analysis of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) were performed on pyroptosis-related differentially expressed genes (PRDEGs) of GSE202659.The key modules for CIRI and pyroptosis were identified by Weight Gene Co-expression Network Analysis (WGCNA). Subsequently, Protein-protein Interaction (PPI) network and the Cytoscape was constructed to screen out hub genes. Used the starBase to predict miRNA interacting with hub genes and constructed mRNA-miRNA-lncRNA interaction networks. CIRI-related Molecular Subtypes were constructed for hub genes. The relationship between immune cells and hub genes was verified via CIBERSORT. Finally, we selected C57BL/6 mice to construct models to confirm hub genes by enzyme linked immunosorbent assay (ELISA), reverse transcription-polymerase chain reaction (RT-PCR), western blot, and Immunofluorescence. RESULTS A total of 272 PRGs and 35 PRDEGs were screened. An eight-gene risk prediction models were established (AUC = 0.868). GO, KEGG, GSEA and GSVA analyses revealed that PRDEGs were mainly involved in positive regulation of cytokine production, and NOD-like receptor signaling pathway. And then, seven hub genes (Irf1, Icam1, Tlr2, Tnf, Cebpb, Il1rn, and Casp8) were identified by PPI. Icam1, Tnf, Cebpb, Il1rn, and Casp8 had high expression profiles in Cluster2 by hierarchical clustering. The immune infiltration analysis results showed that among the hub genes, Cebpb, Il1rn, and Casp8, showed a significant positive correlation with the degree of NK.Actived, and Icam1 showed a significant negative correlation with B.Cells.Memory. The results of animal experiments significantly demonstrated an upregulation of Irf1, Icam1, Tlr2, Cebpb, and Il1rn. CONCLUSION Our finding indicated that Irf1, Icam1, Tlr2, Cebpb, and Il1rn are hub genes associated with pyroptosis, and these genes are all associated with different immune cells, so as to provide new targets for the prevention and treatment of IS from the perspective of pyroptosis.
Collapse
Affiliation(s)
- Xinying Shang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Rui Wei
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Di Yang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Bawei Yu
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Wei Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China.
| |
Collapse
|
4
|
Wang L, Zhang Y, Ji F, Si Z, Liu C, Wu X, Wang C, Chang H. Identification of crucial genes for polycystic ovary syndrome and atherosclerosis through comprehensive bioinformatics analysis and machine learning. Int J Gynaecol Obstet 2025. [PMID: 39981695 DOI: 10.1002/ijgo.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVE To identify potential biomarkers in patients with polycystic ovary syndrome (PCOS) and atherosclerosis, and to explore the common pathologic mechanisms between these two diseases in response to the increased risk of cardiovascular diseases in patients with PCOS. METHODS PCOS and atherosclerosis data sets were downloaded from the GEO database, and their differentially expressed genes were identified. Weighted gene co-expression network analysis was used to obtain intersection genes, and then protein-protein interaction and functional enrichment analysis were performed. Machine learning algorithms were used to select the key genes, which were then validated through external data sets. We constructed a nomogram to predict the risk of atherosclerosis in women with PCOS. Finally, the CIBERSORT algorithm was used to analyze the infiltration of immune cells in the atherosclerosis group. RESULTS We identified six hub genes (CD163, LAPTM5, TNFSF13B, MS4A4A, FGR, and IRF1) that exhibited excellent diagnostic value in validation data sets. Gene ontology terms and KEGG signaling pathway analysis revealed that key genes were associated with immune responses and inflammatory reactions. Abnormal immune cell infiltration was also found in the atherosclerosis group and was correlated with the six hub genes. CONCLUSION Common therapeutic targets of PCOS and atherosclerosis were preliminarily identified through bioinformatics analysis and machine learning techniques. These findings provide new treatment ideas for reducing the risk that PCOS will develop into atherosclerosis.
Collapse
Affiliation(s)
- Lirong Wang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yanli Zhang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fan Ji
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhenmin Si
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengdong Liu
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoke Wu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Obstetrics and Gynecology, Heilongjiang Provincial Hospital, Harbin, China
| | - Chichiu Wang
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Chang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
5
|
Sun C, Sun H, Wei J, Fan X, Simon SI, Passerini AG. IRF-1 Regulates Mitochondrial Respiration and Intrinsic Apoptosis Under Metabolic Stress through ATP Synthase Ancillary Factor TMEM70. Inflammation 2024:10.1007/s10753-024-02209-w. [PMID: 39641858 DOI: 10.1007/s10753-024-02209-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/07/2024]
Abstract
Mitochondrial dysfunction, which can be caused by metabolic stressors such as oxidized low-density lipoprotein (oxLDL), sensitizes the endothelium to pathological changes. The transcription factor interferon regulatory factor 1 (IRF-1) is a master regulator of inflammation, previously shown to promote oxLDL-induced inflammatory pyroptosis in human aortic endothelial cells (HAEC). However, a presumed role for IRF-1 in regulating the intrinsic apoptotic pathway in response to metabolic stress has not been demonstrated. Here targeted deletion of IRF-1 by siRNA in HAEC aggravated oxLDL-induced, mitochondria-mediated intrinsic apoptosis, as evidenced by increased Caspase-3 and Caspase-9 activation, and chromosomal DNA breakage. The increased apoptosis was concomitant with accumulation of mitochondrial ROS, decrease in intracellular ATP production and respiratory oxygen consumption, and abnormal mitochondrial structure. RNA profiling of endothelial cells isolated from wild type and Irf1 knockout mice, followed by quantitative PCR, luciferase activity assay and chromatin immunoprecipitation (ChIP), revealed that IRF-1 directly regulated the expression of transmembrane protein 70 (TMEM70), an ancillary factor required for the assembly of ATP synthase and conversion of an electrochemical gradient to ATP synthesis. Mirroring the effect of IRF1 knockdown, depletion of TMEM70 in HAEC resulted in impaired mitochondrial function and enhanced cell apoptosis. In contrast, overexpression of TMEM70 rescued ATP biosynthesis and suppressed apoptosis in oxLDL-treated, IRF-1-deficient HAEC. These results reveal a novel homeostatic role for IRF-1 in the regulation of mitochondrial function and associated stress-induced apoptosis.
Collapse
Affiliation(s)
- ChongXiu Sun
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, People's Republic of China.
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, People's Republic of China.
| | - Haotian Sun
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, People's Republic of China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, People's Republic of China
| | - Jiahao Wei
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, People's Republic of China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, People's Republic of China
| | - Xing Fan
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, People's Republic of China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, People's Republic of China
| | - Scott I Simon
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| |
Collapse
|
6
|
Shamas S, Rahil RR, Kaushal L, Sharma VK, Wani NA, Qureshi SH, Ahmad SF, Attia SM, Zargar MA, Hamid A, Bhat OM. Pyroptosis in Endothelial Cells and Extracellular Vesicle Release in Atherosclerosis via NF-κB-Caspase-4/5-GSDM-D Pathway. Pharmaceuticals (Basel) 2024; 17:1568. [PMID: 39770410 PMCID: PMC11677252 DOI: 10.3390/ph17121568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Pyroptosis, an inflammatory cell death, is involved in the progression of atherosclerosis. Pyroptosis in endothelial cells (ECs) and its underlying mechanisms in atherosclerosis are poorly understood. Here, we investigated the role of a caspase-4/5-NF-κB pathway in pyroptosis in palmitic acid (PA)-stimulated ECs and EVs as players in pyroptosis. Methods: Human umbilical vein endothelial cells (HUVECs) were cultured in an endothelial cell medium, treated with Ox-LDL, PA, caspase-4/5 inhibitor, NF-κB inhibitor, and sEV release inhibitor for 24 h, respectively. The cytotoxicity of PA was determined using an MTT assay, cell migration using a scratch-wound-healing assay, cell morphology using bright field microscopy, and lipid deposition using oil red O staining. The mRNA and protein expression of GSDM-D, CASP4, CASP5, NF-κB, NLRP3, IL-1β, and IL-18 were determined with RT-PCR and Western blot. Immunofluorescence was used to determine NLRP3 and ICAM-1 expressions. Extracellular vesicles (EVs) were isolated using an exosome isolation kit and were characterized by Western blot and scanning electron microscopy. Results: PA stimulation significantly changed the morphology of the HUVECs characterized by cell swelling, plasma membrane rupture, and increased LDH release, which are features of pyroptosis. PA significantly increased lipid accumulation and reduced cell migration. PA also triggered inflammation and endothelial dysfunction, as evidenced by NLRP3 activation, upregulation of ICAM-1 (endothelial activation marker), and pyroptotic markers (NLRP3, GSDM-D, IL-1β, IL-18). Inhibition of caspase-4/5 (Ac-FLTD-CMK) and NF-κB (trifluoroacetate salt (TFA)) resulted in a significant reduction in LDH release and expression of caspase-4/5, NF-κB, and gasdermin D (GSDM-D) in PA-treated HUVECs. Furthermore, GW4869, an exosome release inhibitor, markedly reduced LDH release in PA-stimulated HUVECs. EVs derived from PA-treated HUVECs exacerbated pyroptosis, as indicated by significantly increased LDH release and augmented expression of GSDM-D, NF-κB. Conclusions: The present study revealed that inflammatory, non-canonical caspase-4/5-NF-κB signaling may be one of the crucial mechanistic pathways associated with pyroptosis in ECs, and pyroptotic EVs facilitated pyroptosis in normal ECs during atherosclerosis.
Collapse
Affiliation(s)
- Salman Shamas
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Razia Rashid Rahil
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Laveena Kaushal
- Department of Dermatology, Venereology & Leprology, Postgraduate Institute for Medical Education and Research, Chandigarh 160012, India; (L.K.); (V.K.S.)
| | - Vinod Kumar Sharma
- Department of Dermatology, Venereology & Leprology, Postgraduate Institute for Medical Education and Research, Chandigarh 160012, India; (L.K.); (V.K.S.)
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Shabir H. Qureshi
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.F.A.); (S.M.A.)
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.F.A.); (S.M.A.)
| | - Mohammad Afzal Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| | - Owais Mohmad Bhat
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India; (S.S.); (R.R.R.); (N.A.W.); (S.H.Q.); (M.A.Z.)
| |
Collapse
|
7
|
Baeza C, Ribagorda M, Maya-Lopez C, Fresno M, Sanchez-Diaz T, Pintor-Chocano A, Sanz AB, Carrasco S, Ortiz A, Sanchez-Niño MD. NIK Is a Mediator of Inflammation and Intimal Hyperplasia in Endothelial Denudation-Induced Vascular Injury. Int J Mol Sci 2024; 25:11473. [PMID: 39519026 PMCID: PMC11546836 DOI: 10.3390/ijms252111473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/27/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Neointimal hyperplasia is the main cause of vascular graft failure in the medium term. NFκB is a key mediator of inflammation that is activated during neointimal hyperplasia following endothelial injury. However, the molecular mechanisms involved in NFκB activation are poorly understood. NFκB may be activated through canonical (transient) and non-canonical (persistent) pathways. NFκB-inducing kinase (NIK, MAP3K14) is the upstream kinase of the non-canonical pathway. We have now explored the impact of NIK deficiency on neointimal hyperplasia following guidewire-induced endothelial cell injury and on local inflammation by comparing NIK activity-deficient alymphoplasia mice (NIKaly/aly) with control wild-type (NIK+/+) mice. Guidewire-induced endothelial cell injury caused neointimal hyperplasia and luminal stenosis and upregulated the local expression of NIK and the NFκB target chemokines monocyte chemoattractant protein-1 (MCP-1/CCL2) and chemokine ligand 5 (RANTES/CCL5). Immunohistochemistry disclosed the infiltration of the media and intima by F4/80 positive macrophages. The intima/media ratio and percentage of stenosis were milder in the NIKaly/aly than in the NIK+/+ mice. Additionally, the gene expression for MCP-1 and RANTES was lower and F4/80+ cell infiltration was milder in the NIKaly/aly than in the NIK+/+ mice. Finally, circulating MCP-1 levels were lower in the NIKaly/aly than in the NIK+/+ mice, reflecting milder systemic inflammation. In conclusion, NIK is a driver of vascular wall inflammation and stenosis following guidewire-induced endothelial cell injury. NIK targeting may be a novel therapeutic approach to limit arterial stenosis following endothelial cell injury.
Collapse
Affiliation(s)
- Ciro Baeza
- Department of Vascular Surgery, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain
| | - Marta Ribagorda
- RICORS2040, 28040 Madrid, Spain
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
| | - Carla Maya-Lopez
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas de la Universidad Autonoma de Madrid, 28049 Madrid, Spain;
| | - Tania Sanchez-Diaz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Aranzazu Pintor-Chocano
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
| | - Ana B. Sanz
- RICORS2040, 28040 Madrid, Spain
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
| | - Susana Carrasco
- RICORS2040, 28040 Madrid, Spain
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
| | - Alberto Ortiz
- RICORS2040, 28040 Madrid, Spain
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- RICORS2040, 28040 Madrid, Spain
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, 28040 Madrid, Spain (A.P.-C.)
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
8
|
Wang L, Zhu Y, Zhang N, Xian Y, Tang Y, Ye J, Reza F, He G, Wen X, Jiang X. The multiple roles of interferon regulatory factor family in health and disease. Signal Transduct Target Ther 2024; 9:282. [PMID: 39384770 PMCID: PMC11486635 DOI: 10.1038/s41392-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024] Open
Abstract
Interferon Regulatory Factors (IRFs), a family of transcription factors, profoundly influence the immune system, impacting both physiological and pathological processes. This review explores the diverse functions of nine mammalian IRF members, each featuring conserved domains essential for interactions with other transcription factors and cofactors. These interactions allow IRFs to modulate a broad spectrum of physiological processes, encompassing host defense, immune response, and cell development. Conversely, their pivotal role in immune regulation implicates them in the pathophysiology of various diseases, such as infectious diseases, autoimmune disorders, metabolic diseases, and cancers. In this context, IRFs display a dichotomous nature, functioning as both tumor suppressors and promoters, contingent upon the specific disease milieu. Post-translational modifications of IRFs, including phosphorylation and ubiquitination, play a crucial role in modulating their function, stability, and activation. As prospective biomarkers and therapeutic targets, IRFs present promising opportunities for disease intervention. Further research is needed to elucidate the precise mechanisms governing IRF regulation, potentially pioneering innovative therapeutic strategies, particularly in cancer treatment, where the equilibrium of IRF activities is of paramount importance.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanghui Zhu
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yali Xian
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Tang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Ye
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fekrazad Reza
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Wen
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Hu H, Wang S, Chen C. Pathophysiological role and potential drug target of NLRP3 inflammasome in the metabolic disorders. Cell Signal 2024; 122:111320. [PMID: 39067838 DOI: 10.1016/j.cellsig.2024.111320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
NLRP3 plays a role in the development of autoinflammatory diseases. NLRP3, ASC, and Caspases 1 or 8 make up the NLRP3 inflammasome, which is an important part of innate immune system. The NLRP3 inflammasome-mediated inflammatory cytokines may also participate in metabolic disorders, such as diabetes, hyperlipidemia, atherosclerosis, non-alcoholic fatty liver disease, and gout. Hence, an overview of the NLRP3 regulation in these metabolic diseases and the potential drugs targeting NLRP3 is the focus of this review.
Collapse
Affiliation(s)
- Huiming Hu
- School of pharmacy, Nanchang Medical College, Nanchang, Jiangxi, China; School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia; Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Jiangxi, China
| | - Shuwen Wang
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia.
| |
Collapse
|
10
|
Hara MA, Ramadan M, Abdelhameid MK, Taher ES, Mohamed KO. Pyroptosis and chemical classification of pyroptotic agents. Mol Divers 2024:10.1007/s11030-024-10987-6. [PMID: 39316325 DOI: 10.1007/s11030-024-10987-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024]
Abstract
Pyroptosis, as a lytic-inflammatory type of programmed cell death, has garnered considerable attention due to its role in cancer chemotherapy and many inflammatory diseases. This review will discuss the biochemical classification of pyroptotic inducers according to their chemical structure, pyroptotic mechanism, and cancer type of these targets. A structure-activity relationship study on pyroptotic inducers is revealed based on the surveyed pyroptotic inducer chemotherapeutics. The shared features in the chemical structures of current pyroptotic inducer agents were displayed, including an essential cyclic head, a vital linker, and a hydrophilic tail that is significant for π-π interactions and hydrogen bonding. The presented structural features will open the way to design new hybridized classes or scaffolds as potent pyroptotic inducers in the future, which may represent a solution to the apoptotic-resistance dilemma along with synergistic chemotherapeutic advantage.
Collapse
Affiliation(s)
- Mohammed A Hara
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al Azhar University (Assiut), Assiut, 71524, Egypt
| | - Mohamed Ramadan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al Azhar University (Assiut), Assiut, 71524, Egypt.
| | - Mohammed K Abdelhameid
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ehab S Taher
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al Azhar University (Assiut), Assiut, 71524, Egypt
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Khaled O Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sinai University (Arish Branch), ElArich, Egypt
| |
Collapse
|
11
|
Wang Y, Chen J, Zheng Y, Jiang J, Wang L, Wu J, Zhang C, Luo M. Glucose metabolite methylglyoxal induces vascular endothelial cell pyroptosis via NLRP3 inflammasome activation and oxidative stress in vitro and in vivo. Cell Mol Life Sci 2024; 81:401. [PMID: 39269632 PMCID: PMC11399538 DOI: 10.1007/s00018-024-05432-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
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.
Collapse
Affiliation(s)
- Yanan Wang
- Basic Medicine Research Innovation Center for Cardiometabolic DiseasesMinistry of EducationLaboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Cancer Early Detection and Treatment Center (CEDTC) and Translational Medicine Research Center (TMRC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
| | - Jinxiang Chen
- Basic Medicine Research Innovation Center for Cardiometabolic DiseasesMinistry of EducationLaboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Youkun Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic DiseasesMinistry of EducationLaboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Liqun Wang
- Basic Medicine Research Innovation Center for Cardiometabolic DiseasesMinistry of EducationLaboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Jianbo Wu
- Basic Medicine Research Innovation Center for Cardiometabolic DiseasesMinistry of EducationLaboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China
| | - Chunxiang Zhang
- Basic Medicine Research Innovation Center for Cardiometabolic DiseasesMinistry of EducationLaboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic DiseasesMinistry of EducationLaboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
- Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China.
| |
Collapse
|
12
|
Wu Z, Li W, Wang S, Zheng Z. Role of deubiquitinase USP47 in cardiac function alleviation and anti-inflammatory immunity after myocardial infarction by regulating NLRP3 inflammasome-mediated pyroptotic signal pathways. Int Immunopharmacol 2024; 136:112346. [PMID: 38850785 DOI: 10.1016/j.intimp.2024.112346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024]
Abstract
Myocardial infarction (MI) is an event of heart attack due to the formation of plaques in the interior walls of the arteries. This study is conducted to explore the role of ubiquitin-specific peptidase 47 (USP47) in cardiac function and inflammatory immunity. MI mouse models were established, followed by an appraisal of cardiac functions, infarct size, pathological changes, and USP47 and NLRP3 levels. MI cell models were established in HL-1 cells using anoxia. Levels of cardiac function-associated proteins, USP7, interferon regulatory factor 1 (IRF1), platelet factor-4 (CXCL4), pyroptotic factors, and neutrophil extracellular traps (NETs) were determined. The bindings of IRF1 to USP47 and the CXCL4 promoter and the ubiquitination of IRF1 were analyzed. USP47 was upregulated in myocardial tissues of MI mice. USP47 inhibition alleviated cardiac functions, and decreased infarct size, pro-inflammatory cytokines, NETs, NLRP3, and pyroptosis. The ubiquitination and expression levels of IRF1 were increased by silencing USP47, and IRF1 bound to the CXCL4 promoter to promote CXCL4. Overexpression of IRF1 or CXCL4 in vitro and injection of Nigericin in vivo reversed the effect of silencing USP47 on alleviating pyroptosis and cardiac functions. Collectively, USP47 stabilized IRF1 and promoted CXCL4, further promoting pyroptosis, impairing cardiac functions, and aggravating immune inflammation through NLRP3 pathways.
Collapse
Affiliation(s)
- Zheng Wu
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Wenzheng Li
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shaoping Wang
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ze Zheng
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Xie J, Yang Y, Zhuo A, Gao M, Tang L, Xiao Y, Zhu H, Fu X. Exosomes derived from mesenchymal stem cells attenuate NLRP3-related pyroptosis in autoimmune premature ovarian insufficiency via the NF-κB pathway. Reprod Biomed Online 2024; 48:103814. [PMID: 38569224 DOI: 10.1016/j.rbmo.2024.103814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/25/2023] [Accepted: 01/04/2024] [Indexed: 04/05/2024]
Abstract
RESEARCH QUESTION What is the effect of exosomes derived from bone marrow mesenchymal stem cells (MSC-Exos) on the pyroptosis and recovery of granulosa cells in autoimmune premature ovarian insufficiency (POI)? DESIGN In vitro, KGN cells were exposed to interferon-gamma to simulate immune injury. Samples were collected after a 48 h incubation with MSC-Exos (30 μg/ml). The cell viability, secretion of oestrogen and expression of key molecules in pyroptosis and the nuclear factor kappa B (NF-κB) pathway were tested. In vivo, the BALB/c mouse model of autoimmune POI model induced by zona pellucida glycoprotein 3 was used. Fertility testing and sample collection were applied 4 weeks after the ovarian subcapsular injection of MSC-Exos (150 μg for each ovary). Hormone concentration measurements, follicle counting and pyroptotic pathway analyses were conducted for each group. RESULTS In vitro, MSC-Exos significantly promoted the proliferation rate and secretion of oestrogen, while at the same time suppressing apoptosis and pyroptosis. In vivo, exosomal treatment normalized the irregular oestrous cycles, rescued the follicular loss and increased the pregnancy rate and number of offspring in POI mice. Elevated serum concentrations of oestrogen and anti-Müllerian hormone, as well as decreased concentrations of FSH and interleukin-1β, were shown. Furthermore, MSC-Exos down-regulated the expression of the NLRP3/Casp1/GSDMD pathway and inhibited activation of the NF-κB pathway. CONCLUSIONS These findings demonstrate for the first time that MSC-Exos exert a significant effect on restoring ovarian function in autoimmune POI in vivo and in vitro by suppressing the NLRP3/Casp1/GSDMD pathway and pyroptosis. The NF-κB pathway may contribute to the regulation of NLRP3-related pyroptosis.
Collapse
Affiliation(s)
- Jiaxin Xie
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China
| | - Yutao Yang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China
| | - Aiping Zhuo
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China
| | - Meng Gao
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China
| | - Lichao Tang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China
| | - Yuanling Xiao
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China
| | - Honglei Zhu
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China
| | - Xiafei Fu
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guanazhou, Guangdong, China.
| |
Collapse
|
14
|
Zhou T, Long K, Chen J, Zhi L, Zhou X, Gao P. Global research progress of endothelial cells and ALI/ARDS: a bibliometric analysis. Front Physiol 2024; 15:1326392. [PMID: 38774649 PMCID: PMC11107300 DOI: 10.3389/fphys.2024.1326392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/18/2024] [Indexed: 05/24/2024] Open
Abstract
Background Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe respiratory conditions with complex pathogenesis, in which endothelial cells (ECs) play a key role. Despite numerous studies on ALI/ARDS and ECs, a bibliometric analysis focusing on the field is lacking. This study aims to fill this gap by employing bibliometric techniques, offering an overarching perspective on the current research landscape, major contributors, and emerging trends within the field of ALI/ARDS and ECs. Methods Leveraging the Web of Science Core Collection (WoSCC) database, we conducted a comprehensive search for literature relevant to ALI/ARDS and ECs. Utilizing Python, VOSviewer, and CiteSpace, we performed a bibliometric analysis on the corpus of publications within this field. Results This study analyzed 972 articles from 978 research institutions across 40 countries or regions, with a total of 5,277 authors contributing. These papers have been published in 323 different journals, spanning 62 distinct research areas. The first articles in this field were published in 2011, and there has been a general upward trend in annual publications since. The United States, Germany, and China are the principal contributors, with Joe G. N. Garcia from the University of Arizona identified as the leading authority in this field. American Journal of Physiology-Lung Cellular and Molecular Physiology has the highest publication count, while Frontiers in Immunology has been increasingly focusing on this field in recent years. "Cell Biology" stands as the most prolific research area within the field. Finally, this study identifies endothelial glycocalyx, oxidative stress, pyroptosis, TLRs, NF-κB, and NLRP3 as key terms representing research hotspots and emerging frontiers in this field. Conclusion This bibliometric analysis provides a comprehensive overview of the research landscape surrounding ALI/ARDS and ECs. It reveals an increasing academic focus on ALI/ARDS and ECs, particularly in the United States, Germany, and China. Our analysis also identifies several emerging trends and research hotspots, such as endothelial glycocalyx, oxidative stress, and pyroptosis, indicating directions for future research. The findings can guide scholars, clinicians, and policymakers in targeting research gaps and setting priorities to advance the field.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kunlan Long
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Chen
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijia Zhi
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiujuan Zhou
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Zhu C, Xu S, Jiang R, Yu Y, Bian J, Zou Z. The gasdermin family: emerging therapeutic targets in diseases. Signal Transduct Target Ther 2024; 9:87. [PMID: 38584157 PMCID: PMC10999458 DOI: 10.1038/s41392-024-01801-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
The gasdermin (GSDM) family has garnered significant attention for its pivotal role in immunity and disease as a key player in pyroptosis. This recently characterized class of pore-forming effector proteins is pivotal in orchestrating processes such as membrane permeabilization, pyroptosis, and the follow-up inflammatory response, which are crucial self-defense mechanisms against irritants and infections. GSDMs have been implicated in a range of diseases including, but not limited to, sepsis, viral infections, and cancer, either through involvement in pyroptosis or independently of this process. The regulation of GSDM-mediated pyroptosis is gaining recognition as a promising therapeutic strategy for the treatment of various diseases. Current strategies for inhibiting GSDMD primarily involve binding to GSDMD, blocking GSDMD cleavage or inhibiting GSDMD-N-terminal (NT) oligomerization, albeit with some off-target effects. In this review, we delve into the cutting-edge understanding of the interplay between GSDMs and pyroptosis, elucidate the activation mechanisms of GSDMs, explore their associations with a range of diseases, and discuss recent advancements and potential strategies for developing GSDMD inhibitors.
Collapse
Affiliation(s)
- Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ruoyu Jiang
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
16
|
Zhang D, Wu H, Liu D, Ye M, Li Y, Zhou G, Yang Q, Liu Y, Li Y. cFLIP L alleviates myocardial ischemia-reperfusion injury by regulating pyroptosis. Cell Biol Int 2024; 48:60-75. [PMID: 37750485 DOI: 10.1002/cbin.12091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/04/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023]
Abstract
Alleviating myocardial ischemia-reperfusion injury (MIRI) plays a critical role in the prognosis and improvement of cardiac function following acute myocardial infarction. Pyroptosis is a newly identified form of cell death that has been implicated in the regulation of MIRI. In our study, H9c2 cells and SD rats were transfected using a recombinant adenovirus vector carrying cFLIPL , and the transfection was conducted for 3 days. Subsequently, H9c2 cells were subjected to 4 h of hypoxia followed by 12 h of reoxygenation to simulate an in vitro ischemia-reperfusion model. SD rats underwent 30 min of ischemia followed by 2 h of reperfusion to establish an MIRI model. Our findings revealed a notable decrease in cFLIPL expression in response to ischemia/reperfusion (I/R) and hypoxia/reoxygenation (H/R) injuries. Overexpression of cFLIPL can inhibit pyroptosis, reducing myocardial infarction area in vivo, and enhancing H9c2 cell viability in vitro. I/R and H/R injuries induced the upregulation of ASC, cleaved Caspase 1, NLRP3, GSDMD-N, IL-1β, and IL-18 proteins, promoting cell apoptosis. Our research indicates that cFLIPL may suppress pyroptosis by strategically binding with Caspase 1, inhibiting the release of inflammatory cytokines and preventing cell membrane rupture. Therefore, cFLIPL could potentially serve as a promising target for alleviating MIRI by suppressing the pyroptotic pathway.
Collapse
Affiliation(s)
- Dong Zhang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - Hui Wu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - Di Liu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - Ming Ye
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - Yunzhao Li
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - Gang Zhou
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - QingZhuo Yang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - YanFang Liu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| | - Yi Li
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
17
|
Katsi V, Papakonstantinou I, Tsioufis K. Atherosclerosis, Diabetes Mellitus, and Cancer: Common Epidemiology, Shared Mechanisms, and Future Management. Int J Mol Sci 2023; 24:11786. [PMID: 37511551 PMCID: PMC10381022 DOI: 10.3390/ijms241411786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The involvement of cardiovascular disease in cancer onset and development represents a contemporary interest in basic science. It has been recognized, from the most recent research, that metabolic syndrome-related conditions, ranging from atherosclerosis to diabetes, elicit many pathways regulating lipid metabolism and lipid signaling that are also linked to the same framework of multiple potential mechanisms for inducing cancer. Otherwise, dyslipidemia and endothelial cell dysfunction in atherosclerosis may present common or even interdependent changes, similar to oncogenic molecules elevated in many forms of cancer. However, whether endothelial cell dysfunction in atherosclerotic disease provides signals that promote the pre-clinical onset and proliferation of malignant cells is an issue that requires further understanding, even though more questions are presented with every answer. Here, we highlight the molecular mechanisms that point to a causal link between lipid metabolism and glucose homeostasis in metabolic syndrome-related atherosclerotic disease with the development of cancer. The knowledge of these breakthrough mechanisms may pave the way for the application of new therapeutic targets and for implementing interventions in clinical practice.
Collapse
Affiliation(s)
- Vasiliki Katsi
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
| | | | - Konstantinos Tsioufis
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|