1
|
Zheng Y, Zhang X, Wang Z, Zhang R, Wei H, Yan X, Jiang X, Yang L. MCC950 as a promising candidate for blocking NLRP3 inflammasome activation: A review of preclinical research and future directions. Arch Pharm (Weinheim) 2024; 357:e2400459. [PMID: 39180246 DOI: 10.1002/ardp.202400459] [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: 06/06/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
The NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome is a key component of the innate immune system that triggers inflammation and pyroptosis and contributes to the development of several diseases. Therefore, blocking the activation of the NLRP3 inflammasome has therapeutic potential for the treatment of these diseases. MCC950, a selective small molecule inhibitor, has emerged as a promising candidate for blocking NLRP3 inflammasome activation. Ongoing research is focused on elucidating the specific targets of MCC950 as well as assessfing its metabolism and safety profile. This review discusses the diseases that have been studied in relation to MCC950, with a focus on stroke, Alzheimer's disease, liver injury, atherosclerosis, diabetes mellitus, and sepsis, using bibliometric analysis. It then summarizes the potential pharmacological targets of MCC950 and discusses its toxicity. Furthermore, it traces the progression from preclinical to clinical research for the treatment of these diseases. Overall, this review provides a solid foundation for the clinical therapeutic potential of MCC950 and offers insights for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Yujia Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Ziyu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Ruifeng Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin, China
| | - Lin Yang
- School of Medicial Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, Jinghai, China
| |
Collapse
|
2
|
Tang F, Zhao XL, Xu LY, Zhang JN, Ao H, Peng C. Endothelial dysfunction: Pathophysiology and therapeutic targets for sepsis-induced multiple organ dysfunction syndrome. Biomed Pharmacother 2024; 178:117180. [PMID: 39068853 DOI: 10.1016/j.biopha.2024.117180] [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: 05/22/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Sepsis and septic shock are critical medical conditions characterized by a systemic inflammatory response to infection, significantly contributing to global mortality rates. The progression to multiple organ dysfunction syndrome (MODS) represents the most severe complication of sepsis and markedly increases clinical mortality. Central to the pathophysiology of sepsis, endothelial cells play a crucial role in regulating microcirculation and maintaining barrier integrity across various organs and tissues. Recent studies have underscored the pivotal role of endothelial function in the development of sepsis-induced MODS. This review aims to provide a comprehensive overview of the pathophysiology of sepsis-induced MODS, with a specific focus on endothelial dysfunction. It also compiles compelling evidence regarding potential small molecules that could attenuate sepsis and subsequent multi-organ damage by modulating endothelial function. Thus, this review serves as an essential resource for clinical practitioners involved in the diagnosing, managing, and providing intensive care for sepsis and associated multi-organ injuries, emphasizing the importance of targeting endothelial cells to enhance outcomes of the patients.
Collapse
Affiliation(s)
- Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
3
|
Luo M, Peng Y, Lv D, Xue Y, Huang L, Hu Y, Zhu W, Luo S, Shen J, Li X. LncRNA GAS5 downregulates NLRP3 inflammasome activation-mediated pyroptosis in sepsis-induced myocardial injury by targeting SIRT3/AMPKα. Heliyon 2023; 9:e22939. [PMID: 38076153 PMCID: PMC10703703 DOI: 10.1016/j.heliyon.2023.e22939] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2024] Open
Abstract
An increasing body of studies has demonstrated the significance of long non-coding RNA (lncRNA) growth arrest specific 5 (GAS5) in inflammation and myocardial injury in septic shock. This research aims to determine whether GAS5 contributes to the pathological development of sepsis-induced cardiac damage and NLRP3 inflammasome-mediated myocardial cell pyroptosis. Cecal ligation and puncture (CLP) surgery was used to cause septic shock in C57BL/6 wild-type mice. After CLP, inflammatory, pyroptosis parameters of myocardial tissue, survival rate, and Murine Sepsis Score (MSS) were assessed to evaluate the involvement of GAS5 in the mouse myocardial depression. To investigate GAS5's function in lipopolysaccharide (LPS) induced myocardial cell pyroptosis, gain- and loss-of-function experiments were conducted in vitro on HL-1 cells. Our findings indicated that CLP dramatically reduced survival rates, MSS, SIRT3 and p-AMPK expression, and activated the Nuclear factor-κB (NF-κB) pathway and NLRP3 inflammasome-mediated pyroptosis. The NF-κB and pyroptosis pathways were greatly elevated while SIRT3/p-AMPKα was dramatically decreased as a result of GAS5 being downregulated. Meanwhile, the regulatory effect could be suppressed by SIRT3 and AMPKα activator. Our observations supported the idea that GAS5 has a crucial protective impact against myocardial inflammation and pyroptosis in sepsis.
Collapse
Affiliation(s)
- Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, 400016, Chongqing, China
| | - Yuce Peng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, 400016, Chongqing, China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, 400016, Chongqing, China
| | - Yuzhou Xue
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Longxiang Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Hu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, 400016, Chongqing, China
| | - Wenyan Zhu
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Medical Data Research Institute of Chongqing Medical University, Chongqing Medical University, Chongqing, 400016, China
| | - Jian Shen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiang Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
4
|
Ibadi MH, Majeed S, Ghafil FA, Hadi NR. Regorafenib modulation of the angiopoietin/TIE2 axis in a mouse model of sepsis-induced lung injury. J Med Life 2023; 16:1639-1645. [PMID: 38406775 PMCID: PMC10893570 DOI: 10.25122/jml-2023-0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/07/2023] [Indexed: 02/27/2024] Open
Abstract
Sepsis, often resulting from an immune response overreaction to microorganisms and their products, can lead to acute lung injury through inflammation mediated by excessive cytokines. This study aimed to investigate the effects of regorafenib on lung injury in mice following the induction of sepsis. We divided mice into four groups (n=6 each): a sham group (undergoing laparotomy without cecal ligation and puncture [CLP]), a CLP group, a vehicle group, and a regorafenib-treated group (30 mg/kg IP, administered one hour before CLP). TNF-α, IL-1β, VEGF, MPO, caspase-11, and Ang-2 levels were significantly increased (p<0.05) in the CLP group compared to the sham group, while the regorafenib group showed significant reductions in these markers versus the CLP group (p< 0.05). In contrast, Ang-1 levels, which were reduced in the CLP group (p<0.05) compared to the sham group, were elevated in the regorafenib group compared to the CLP group. Quantitative real-time PCR revealed a significant decrease in TIE2 and VE-cadherin mRNA expression in the lung tissue of the CLP group compared to the sham group. There were no significant differences in mRNA expression of the TIE2 gene between the regorafenib and CLP group. However, VE-cadherin significantly increased after regorafenib treatment. Regorafenib demonstrated lung-protective effects through its anti-inflammatory and antiangiogenic activities and its influence on lung tissue mRNA expression of the cadherin gene.
Collapse
Affiliation(s)
| | - Sahar Majeed
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Fadhaa Abdulameer Ghafil
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| |
Collapse
|
5
|
Wang Y, Wu J, Wang J, He L, Lai H, Zhang T, Wang X, Li W. Mitochondrial oxidative stress in brain microvascular endothelial cells: Triggering blood-brain barrier disruption. Mitochondrion 2023; 69:71-82. [PMID: 36709855 DOI: 10.1016/j.mito.2023.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/02/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Blood-brain barrier disruption plays an important role in central nervous system diseases. This review provides information on the role of mitochondrial oxidative stress in brain microvascular endothelial cells in cellular dysfunction, the disruption of intercellular junctions, transporter dysfunction, abnormal angiogenesis, neurovascular decoupling, and the involvement and aggravation of vascular inflammation and illustrates related molecular mechanisms. In addition, recent drug and nondrug therapies targeting cerebral vascular endothelial cell mitochondria to repair the blood-brain barrier are discussed. This review shows that mitochondrial oxidative stress disorder in brain microvascular endothelial cells plays a key role in the occurrence and development of blood-brain barrier damage and may be critical in various pathological mechanisms of blood-brain barrier damage. These new findings suggest a potential new strategy for the treatment of central nervous system diseases through mitochondrial modulation of cerebral vascular endothelial cells.
Collapse
Affiliation(s)
- Yi Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jing Wu
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jiexin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Linxi He
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Han Lai
- School of Foreign Languages, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Tian Zhang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Xin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| |
Collapse
|
6
|
PCSK9 Promotes Endothelial Dysfunction During Sepsis Via the TLR4/MyD88/NF-κB and NLRP3 Pathways. Inflammation 2023; 46:115-128. [PMID: 35930089 DOI: 10.1007/s10753-022-01715-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/05/2022]
Abstract
Endothelial dysfunction often accompanies sepsis. We aimed to explore the role of PCSK9 in septic endothelial dysfunction. Sepsis was induced by lipopolysaccharide (LPS) treatment of human umbilical vein endothelial cells (HUVECs) in vitro and cecal ligation and puncture (CLP) surgery in mice in vivo. Evolocumab (EVC) and Pep 2-8, PCSK9 inhibitors, were subsequently used to determine the role of PCSK9 in sepsis-induced endothelial dysfunction in vitro and in vivo, respectively. In addition, the TLR4 agonist, Kdo2-Lipid A ammonium (KLA), was used to determine the related mechanism. Protein expression of eNOS, VE-cadherin, PCSK9, TLR4, MyD88, p-p65, p65, NLRP3, ASC, and caspase-1 p20 in mice aortas and HUVECs was measured by western blotting, while mRNA expression of TNFα, IL-1β, and IL-18 was determined by qRT-PCR. The level of inflammatory cytokines of mouse aortas was visualized by immunohistochemistry. Vasodilation of the aorta was detected by vascular reactivity experiments. The 96-h survival rate after CLP was assessed. Our findings showed that the expression of eNOS and VE-cadherin decreased, and PCSK9 expression increased, in septic HUVECs or mice. Inhibition of PCSK9 increased eNOS and VE-cadherin expression. Activation of the TLR4/MyD88/NF-κB and NLRP3 pathways may be responsible for PCSK9-induced endothelial dysfunction in sepsis. Vascular reactivity tests and survival studies showed that inhibition of PCSK9 could prevent the decrease in endothelium-dependent vasodilation function and improve the survival rates of septic mice. In summary, our results suggested that increased PCSK9 expression during sepsis activated the TLR4/MyD88/NF-κB and NLRP3 pathways to induce inflammation, which resulted in vascular endothelial dysfunction and decreased survival rates. Thus, inhibition of PCSK9 may be a potential clinical therapeutic target to improve vascular endothelial function in sepsis.
Collapse
|
7
|
Luo M, Luo S, Xue Y, Chang Q, Yang H, Dong W, Zhang T, Cao S. Aerobic exercise inhibits renal EMT by promoting irisin expression in SHR. iScience 2023; 26:105990. [PMID: 36798442 PMCID: PMC9926087 DOI: 10.1016/j.isci.2023.105990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/03/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
To determine the effect of aerobic exercise in different intensities on renal injury and epithelial-mesenchymal transformation (EMT) in the kidney of spontaneously hypertensive rats (SHR) and explore possible mechanisms, we subjected SHR to different levels of 14-week aerobic treadmill training. We tested the effects of aerobic exercise on irisin level, renal function, and EMT modulators in the kidney. We also treated angiotensin II-induced HK-2 cells with irisin and tested the changes in EMT levels. The data showed low and moderate aerobic exercise improved renal function and inhibited EMT through promoting irisin expression in SHR. However, high-intensity exercise training had no effect on renal injury and EMT in SHR but did significantly activate STAT3 phosphorylation in the kidney. These results clarify the mechanisms of exercise in improving hypertension-related renal injury and suggest that irisin might be a therapeutic target for patients with kidney injury.
Collapse
Affiliation(s)
- Minghao Luo
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuzhou Xue
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Chang
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| | - Hui Yang
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Wenyu Dong
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Zhang
- The Fifth Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Shuyuan Cao
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
- The Fifth Affiliated Hospital of Sun Yat-sen University, Guangdong, China
- Corresponding author
| |
Collapse
|
8
|
Papadopoulos KI, Papadopoulou A, Aw TC. A protective erythropoietin evolutionary landscape, NLRP3 inflammasome regulation, and multisystem inflammatory syndrome in children. Hum Cell 2023; 36:26-40. [PMID: 36310304 PMCID: PMC9618415 DOI: 10.1007/s13577-022-00819-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2022]
Abstract
The low incidence of pediatric severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection and the associated multisystem inflammatory syndrome (MIS-C) lack a unifying pathophysiological explanation, impeding effective prevention and therapy. Activation of the NACHT, LRR, and PYD domains-containing protein (NLRP) 3 inflammasome in SARS-CoV-2 with perturbed regulation in MIS-C, has been reported. We posit that, early age physiological states and genetic determinants, such as certain polymorphisms of renin-angiotensin aldosterone system (RAAS) molecules, promote a controlled RAAS hyperactive state, and form an evolutionary landscape involving an age-dependent erythropoietin (EPO) elevation, mediating ancestral innate immune defenses that, through appropriate NLRP3 regulation, mitigate tissue injury and pathogen invasion. SARS-CoV-2-induced downregulation of angiotensin-converting enzyme (ACE)2 expression in endothelial cells (EC), impairment of endothelial nitric oxide (NO) synthase (eNOS) activity and downstream NO bioavailability, may promote a hyperactive RAAS with elevated angiotensin II and aldosterone that, can trigger, and accelerate NLRP3 inflammasome activation, while EPO-eNOS/NO abrogate it. Young age and a protective EPO evolutionary landscape may successfully inhibit SARS-CoV-2 and contain NLRP3 inflammasome activation. By contrast, increasing age and falling EPO levels, in genetically susceptible children with adverse genetic variants and co-morbidities, may lead to unopposed RAAS hyperactivity, NLRP3 inflammasome dysregulation, severe endotheliitis with pyroptotic cytokine storm, and development of autoantibodies, as already described in MIS-C. Our haplotype estimates, predicted from allele frequencies in population databases, are in concordance with MIS-C incidence reports in Europeans but indicate lower risks for Asians and African Americans. Targeted Mendelian approaches dissecting the influence of relevant genetic variants are needed.
Collapse
Affiliation(s)
- Konstantinos I Papadopoulos
- Department of Research and Development, THAI StemLife Co., Ltd., 566/3 THAI StemLife Bldg., Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd., Wangthonglang, 10310, Bangkok, Thailand.
| | - Alexandra Papadopoulou
- Occupational and Environmental Health Services, Feelgood Lund, Ideon Science Park, Scheelevägen 17, 223 63, Lund, Sweden
| | - Tar-Choon Aw
- Department of Laboratory Medicine, Changi General Hospital, Singapore, 529889, Singapore
- Department of Medicine, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|
9
|
Yu H, Liao K, Hu Y, Lv D, Luo M, Liu Q, Huang L, Luo S. Role of the cGAS-STING Pathway in Aging-related Endothelial Dysfunction. Aging Dis 2022; 13:1901-1918. [PMID: 36465181 PMCID: PMC9662267 DOI: 10.14336/ad.2022.0316] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/16/2022] [Indexed: 07/30/2023] Open
Abstract
Endothelial dysfunction develops gradually with age, and is the foundation of many age-related diseases in the elderly. The purpose of this study was to investigate the role of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway in aging-related endothelial dysfunction. Endothelial functional parameters and biochemical indices of vascular function were examined in 2-, 6-, 12- and 24-month-old mice. Then, 6-month-old mice were administered RU.521, a specific inhibitor of cGAS, for 6 months, and endothelial functional parameters and biochemical indices of vascular function were re-examined. An in vitro model of cell senescence was established by treating human aortic endothelial cells (HAECs) with D-Galactose (D-GAL). Using inhibitors or siRNA interference, cGAS and STING were suppressed or silenced in senescent HAECs, and changes in the expression of eNOS, the senescence markers, p53, p21 and p16, components of the cGAS-STING pathway and Senescence-Associated β-galactosidase (SA-β-gal) staining were examined. Finally, cGAS, STING and p-IRF3 levels were measured in aorta tissue sections from eight patients. A decline in endothelial function, up-regulation of p53, p21 and p16 expression, and activation of the cGAS-STING pathway were observed in aging mice. Inhibition of cGAS was found to improve endothelial function and reverse the increased expression of aging markers. Our in vitro data demonstrated that D-GAL induced a decrease in eNOS expression and cell senescence, which could be partly reversed by cGAS inhibitor, STING inhibitor, siRNA-cGAS and siRNA-STING treatment. Higher expression levels of cGAS, STING and p-IRF3 were observed in aged human aortic intima tissue compared to young aortic intima tissue. Our study demonstrated that activation of the cGAS-STING pathway played a vital role in aging-related endothelial dysfunction. Thus, the cGAS-STING pathway may be a potential target for the prevention of cardiovascular diseases in the elderly.
Collapse
Affiliation(s)
- Huilin Yu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ke Liao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yu Hu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Qian Liu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Longxiang Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Liu R, Luo X, Li J, Lei Y, Zeng F, Huang X, Lan Y, Yang F. Melatonin: A window into the organ-protective effects of sepsis. Biomed Pharmacother 2022; 154:113556. [PMID: 35994818 DOI: 10.1016/j.biopha.2022.113556] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/22/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022] Open
Abstract
Sepsis is an uncontrolled host response to infection. In some cases, it progresses to multi-organ insufficiency, leading to septic shock and increased risk of mortality. Various organ support strategies are currently applied clinically, but they are still inadequate in terms of reducing mortality. Melatonin is a hormone that regulates sleep and wakefulness, and it is associated with a reduced risk of death in patients with sepsis. Evidence suggests that melatonin may help protect organ function from sepsis-related damage. Here, we review information related to the role of melatonin in protecting organ function during sepsis and explore its potential clinical applications, with the aim of providing an effective therapeutic strategy for treating sepsis-induced organ insufficiency.
Collapse
Affiliation(s)
- Rongan Liu
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiu Luo
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiajia Li
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Lei
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fan Zeng
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaobo Huang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunping Lan
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Fuxun Yang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
11
|
Recurrent Hypoglycemia Impaired Vascular Function in Advanced T2DM Rats by Inducing Pyroptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7812407. [PMID: 35915611 PMCID: PMC9338872 DOI: 10.1155/2022/7812407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/14/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022]
Abstract
Background Hypoglycemia is a dangerous side effect of intensive glucose control in diabetes. Even though it leads to adverse cardiovascular events, the effects of hypoglycemia on vascular biology in diabetes have not been adequately studied. Methods Aged Sprague-Dawley rats were fed a high-fat diet and given streptozotocin to induce type 2 diabetes mellitus (T2DM). Acute and recurrent hypoglycemia were then induced by glucose via insulin administration. Vascular function, oxidative stress, and pyroptosis levels in aortic tissue were assessed by physiological and biochemical methods. Results Hypoglycemia was associated with a marked decrease in vascular function, elevated oxidative stress, and elevated pyroptosis levels in the thoracic aorta. The changes in oxidative stress and pyroptosis were greater in rats with recurrent hypoglycemia than in those with acute hypoglycemia. Conclusions Hypoglycemia impaired vascular function in aged rats with T2DM by inducing pyroptosis. The extent of injury increased with the duration of blood glucose fluctuation.
Collapse
|
12
|
NLRP3 inflammasome contributes to endothelial dysfunction in angiotensin II-induced hypertension in mice. Microvasc Res 2022; 143:104384. [PMID: 35618036 DOI: 10.1016/j.mvr.2022.104384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/26/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022]
Abstract
AIMS Inflammation is a key feature of endothelial dysfunction induced by angiotensin (Ang) II. The purpose of this study was to explore the role of Nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome in endothelial dysfunction in Ang II-induced hypertension. MATERIALS AND METHODS We analyzed blood pressure and vascular function of wild-type (WT) and Nlrp3 knockout (Nlrp3-/-) mice, treated with Ang II. In vitro, we mainly tested the endothelial nitric oxide synthase (eNOS) phosphorylation expression of human umbilical vein endothelial cells (HUVECs). KEY FINDINGS Here we showed that 14-day Ang II infusion into mice resulted in the elevation of blood pressure, NLRP3 expression, serum interleukin (IL)-1β level, and the decline of endothelium-dependent relaxation function, p-eNOS-Ser1177 expression in aortas. Nlrp3 deficiency reduced Ang II-induced blood pressure elevation and endothelial dysfunction. In vitro, NLRP3 was involved in the effect of Ang II on reducing p-eNOS-Ser1177 expression. Moreover, the direct effect of IL-1β on vascular endothelial injury could be observed in both vivo and vitro. SIGNIFICANCE Our result demonstrates that the NLRP3 inflammasome is critically involved in the detrimental effects of Ang II on vascular endothelium in hypertension via the activation of IL-1β, placing NLRP3 as a potential target for therapeutic interventions in conditions with endothelial dysfunction in hypertension.
Collapse
|
13
|
SIRT3-AMPK signaling pathway as a protective target in endothelial dysfunction of early sepsis. Int Immunopharmacol 2022; 106:108600. [PMID: 35217431 DOI: 10.1016/j.intimp.2022.108600] [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: 10/29/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/15/2022]
Abstract
Extensive vascular endothelial dysfunction usually occurs in sepsis, resulting in high mortality. The purpose of this study was therefore to investigate the role of AMP-dependent protein kinase (AMPK) in the aortic endothelial dysfunction of early sepsis in mice, and the relationship between AMPK and Sirtuin3 (SIRT3). Cecal ligation and puncture (CLP) surgery was performed to establish a mouse sepsis model, and human umbilical vein endothelial cells (HUVECs) were treated with lipopolysaccharide (LPS) to mimic a sepsis model in vitro. We suppressed and increased the activities of AMPK with Dorsomorphin (CC) and Acadesine (AICAR), respectively. 3-TYP (SIRT3 inhibitor) and Honokiol (SIRT3 agonist) were used to alter SIRT3 activity. Then, the inflammatory and endothelial function parameters of the vascular tissue and survival rate were determined. In vivo, the expression of Ser1177 phosphorylation of endothelial nitric oxide synthase (p-eNOS), endothelium-dependent relaxation function, and survival decreased (P < 0.05), while NF-κB and NLRP3 pathways were activated in CLP-induced early sepsis (P < 0.05). Moreover, activation of AMPK significantly reversed the reduction of p-eNOS expression (P < 0.05), prevented endothelial dysfunction (P < 0.05), deactivated NF-κB and NLRP3 pathways (P < 0.05), and improved survival (P < 0.05) in septic mice. However, AMPK inhibition led to opposite effects (P < 0.05). In addition, changing the activity of AMPK had little effect on SIRT3 expression (P > 0.05), while the expression of p-AMPK varied with the inhibition or activation of SIRT3 (P < 0.05), which was further demonstrated using in vitro experiments. Together, the results showed that the SIRT3-AMPK signaling pathway played an important role in inhibiting vascular inflammation and endothelial dysfunction during early sepsis.
Collapse
|
14
|
He A, Shen J, Xue Y, Xiang Li, Li Y, Huang L, Lv D, Luo M. Diacerein attenuates vascular dysfunction by reducing inflammatory response and insulin resistance in type 2 diabetic rats. Biochem Biophys Res Commun 2021; 585:68-74. [PMID: 34801936 DOI: 10.1016/j.bbrc.2021.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 12/23/2022]
Abstract
AIM To examine the effect of diacerein on vascular dysfunction in type 2 diabetic rats and elucidate the mechanism of diacerein. METHODS In a rat model, type 2 diabetes was induced by high-fat diet and streptozotocin. Vascular function was assessed in vascular reactivity experiment. The effect of diacerein (10 or 20 mg/kg/day) on blood glucose, inflammation and insulin signaling, and modulators in vascular tissue in diabetic rats were investigated by molecular and biochemical approaches. RESULTS In this study, diacerein inhibited diabetes-induced vascular dysfunction. Diacerein treatment normalized blood glucose, insulin tolerance test, inflammatory cytokine levels and nitric oxide synthases expression in diabetic rats. Moreover, diacerein inhibited NF-κB and NLRP3 pathways and activated insulin signaling pathway related proteins IRS-1 and AKT in diabetic rats. CONCLUSION Diacerein improved vascular function effectively in diabetic rats by suppressing inflammation and reducing insulin resistance. These results suggest that diacerein may represent a novel therapy for patients with diabetes.
Collapse
Affiliation(s)
- An He
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jian Shen
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuzhou Xue
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiang Li
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuanjing Li
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Longxiang Huang
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dingyi Lv
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Minghao Luo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
15
|
Liao K, Lv DY, Yu HL, Chen H, Luo SX. iNOS regulates activation of the NLRP3 inflammasome through the sGC/cGMP/PKG/TACE/TNF-α axis in response to cigarette smoke resulting in aortic endothelial pyroptosis and vascular dysfunction. Int Immunopharmacol 2021; 101:108334. [PMID: 34768128 DOI: 10.1016/j.intimp.2021.108334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cigarette smoke (CS) is associated with vascular injury and dysfunction, which may be mediated by iNOS and NLRP3. However, the exact mechanism is unknown. METHODS iNOS-knockout and NLRP3-knockout C57BL/6 mice were exposed to air or CS. The vascular structure was examined by hematoxylin-eosin staining. The vascular tension was measured by a vascular reactivity assay. The expression of iNOS, NLRP3, caspase-1p20, IL-1β and eNOS were measured by western blotting. Human aortic endothelial cells (HAECs) were exposed to L-NIL (iNOS inhibitor), MCC950 (NLRP3 inhibitor), ODQ (sGC inhibitor), KT5823 (PKG inhibitor) or TAPI-1 (TACE/ADAM17 inhibitor) for 1 h prior to cigarette smoke extract (CSE) treatment. The cell viability and lactate dehydrogenase activity were assessed and pyroptosis was determined by scanning electron microscopy. The mRNA expression of TNF-α, and protein expression of iNOS, active-TACE, NLRP3, caspase-1p20, IL-1β, and eNOS were measured. RESULTS CS resulted in shrinkage of endothelial cells, impaired aorta relaxation, reduced eNOS expression, and induced expression of iNOS, NLRP3, caspase-1p20 and IL-1β, which could be prevented by knockdown of iNOS and NLRP3. CSE reduced cell viability, induced LDH release and pyroptosis, and promoted iNOS, NLRP3, caspase-1p20, and IL-1β expression and reduced eNOS reduction, which could be reversed by inhibition of iNOS or NLRP3 in HAECs. Altogether, activation of the NLRP3 inflammasome by iNOS in CS-exposed HAECs may be mediated by the sGC/cGMP/PKG/TACE/TNF- α pathway. CONCLUSION These results link iNOS to NLRP3 in CSE-stimulated HAECs through the sGC/cGMP/PKG/TACE/TNF-α pathway. The findings identify a mechanism through which iNOS and NLRP3 contribute to the pathogenesis of CS-induced pyroptosis and impaired aorta relaxation in HAECs.
Collapse
Affiliation(s)
- Ke Liao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Ding-Yi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hui-Lin Yu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hong Chen
- Department of Respiratory, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| | - Su-Xin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| |
Collapse
|
16
|
Nunes PR, Mattioli SV, Sandrim VC. NLRP3 Activation and Its Relationship to Endothelial Dysfunction and Oxidative Stress: Implications for Preeclampsia and Pharmacological Interventions. Cells 2021; 10:cells10112828. [PMID: 34831052 PMCID: PMC8616099 DOI: 10.3390/cells10112828] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia (PE) is a specific syndrome of human pregnancy, being one of the main causes of maternal death. Persistent inflammation in the endothelium stimulates the secretion of several inflammatory mediators, activating different signaling patterns. One of these mechanisms is related to NLRP3 activation, initiated by high levels of danger signals such as cholesterol, urate, and glucose, producing IL-1, IL-18, and cell death by pyroptosis. Furthermore, reactive oxygen species (ROS), act as an intermediate to activate NLRP3, contributing to subsequent inflammatory cascades and cell damage. Moreover, increased production of ROS may elevate nitric oxide (NO) catabolism and consequently decrease NO bioavailability. NO has many roles in immune responses, including the regulation of signaling cascades. At the site of inflammation, vascular endothelium is crucial in the regulation of systemic inflammation with important implications for homeostasis. In this review, we present the important role of NLRP3 activation in exacerbating oxidative stress and endothelial dysfunction. Considering that the causes related to these processes and inflammation in PE remain a challenge for clinical practice, the use of drugs related to inhibition of the NLRP3 may be a good option for future solutions for this disease.
Collapse
|
17
|
Luo M, Cao C, Niebauer J, Yan J, Ma X, Chang Q, Zhang T, Huang X, Liu G. Effects of different intensities of continuous training on vascular inflammation and oxidative stress in spontaneously hypertensive rats. J Cell Mol Med 2021; 25:8522-8536. [PMID: 34331512 PMCID: PMC8419160 DOI: 10.1111/jcmm.16813] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 12/17/2022] Open
Abstract
We aimed to study the effects and underlying mechanism of different intensities of continuous training (CT) on vascular inflammation and oxidative stress in spontaneously hypertensive rats (SHR). Rats were divided into five groups (n = 12): Wistar‐Kyoto rats sedentary group (WKY‐S), sedentary group (SHR‐S), low‐intensity CT group (SHR‐L), medium‐intensity CT group (SHR‐M) and high‐intensity CT group (SHR‐H). Changes in body mass, heart rate and blood pressure were recorded. The rats were euthanized after 14 weeks, and blood and vascular tissue samples were collected. Haematoxylin and Eosin staining was used to observe the aortic morphology, and Western blot was used to detect the expression of mesenteric artery proteins. After CT, the mean arterial pressures improved in SHR‐L and SHR‐M and increased in SHR‐H compared with those in SHR‐S. Vascular inflammation and oxidative stress levels significantly subsided in SHR‐L and SHR‐M (p < 0.05), whereas in SHR‐H, only vascular inflammation significantly subsided (p < 0.05), and oxidative stress remained unchanged (p > 0.05). AMPK and SIRT1/3 expressions in SHR‐L and SHR‐M were significantly up‐regulated than those in SHR‐S (p < 0.05). These results indicated that low‐ and medium‐intensity CT can effectively reduce the inflammatory response and oxidative stress of SHR vascular tissue, and high‐intensity CT can improve vascular tissue inflammation but not oxidative stress.
Collapse
Affiliation(s)
- Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunmei Cao
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Josef Niebauer
- University Institute of Sports Medicine, Prevention and Rehabilitation, Paracelsus Medical University, Salzburg, Austria
| | - Jianghong Yan
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xindong Ma
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Qing Chang
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| | - Ting Zhang
- The Fifth Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Xiaoxiao Huang
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| | - Guochun Liu
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Corcoran SE, Halai R, Cooper MA. Pharmacological Inhibition of the Nod-Like Receptor Family Pyrin Domain Containing 3 Inflammasome with MCC950. Pharmacol Rev 2021; 73:968-1000. [PMID: 34117094 DOI: 10.1124/pharmrev.120.000171] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Activation of the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome drives release of the proinflammatory cytokines interleukin (IL)-1β and IL-18 and induces pyroptosis (lytic cell death). These events drive chronic inflammation, and as such, NLRP3 has been implicated in a large number of human diseases. These range from autoimmune conditions, the simplest of which is NLRP3 gain-of-function mutations leading to an orphan disease, cryopyrin-associated period syndrome, to large disease burden indications, such as atherosclerosis, heart failure, stroke, neurodegeneration, asthma, ulcerative colitis, and arthritis. The potential clinical utility of NLRP3 inhibitors is substantiated by an expanding list of indications in which NLRP3 activation has been shown to play a detrimental role. Studies of pharmacological inhibition of NLRP3 in nonclinical models of disease using MCC950 in combination with human genetics, epigenetics, and analyses of the efficacy of biologic inhibitors of IL-1β, such as anakinra and canakinumab, can help to prioritize clinical trials of NLRP3-directed therapeutics. Although MCC950 shows excellent (nanomolar) potency and high target selectivity, its pharmacokinetic and toxicokinetic properties limited its therapeutic development in the clinic. Several improved, next-generation inhibitors are now in clinical trials. Hence the body of research in a plethora of conditions reviewed herein may inform analysis of the potential translational value of NLRP3 inhibition in diseases with significant unmet medical need. SIGNIFICANCE STATEMENT: The nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is one of the most widely studied and best validated biological targets in innate immunity. Activation of NLRP3 can be inhibited with MCC950, resulting in efficacy in more than 100 nonclinical models of inflammatory diseases. As several next-generation NLRP3 inhibitors are entering proof-of-concept clinical trials in 2020, a review of the pharmacology of MCC950 is timely and significant.
Collapse
Affiliation(s)
- Sarah E Corcoran
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Reena Halai
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Matthew A Cooper
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| |
Collapse
|
19
|
You L, Zhang D, Geng H, Sun F, Lei M. Salidroside protects endothelial cells against LPS-induced inflammatory injury by inhibiting NLRP3 and enhancing autophagy. BMC Complement Med Ther 2021; 21:146. [PMID: 34011327 PMCID: PMC8136193 DOI: 10.1186/s12906-021-03307-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 04/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Salidroside (SAL) is a bioactive compound extracted from Rhodiola rosea with various biological properties. This study was designed to explore the functions of SAL on the endothelial damage induced by lipopolysaccharide (LPS) and its related mechanisms. METHODS Human umbilical vein endothelial cells (HUVECs) were pretreated with SAL (0, 10, 25, 50, 100 μM), and then incubated with LPS (10 μg/mL). Cell viability was evaluated by MTT assay, cell injury by lactate dehydrogenase (LDH) release, and inflammatory cytokines release by ELISA assay. Oxidative stress was evaluated by malondialdehyde (MDA) and superoxide dismutase (SOD) in cell lysate. Apoptosis was detected by flow cytometry and caspase-3 activity. Western blot were performed to determine expression levels of autophagy and NOD-like receptor protein 3 (NLRP3) related proteins. RESULTS SAL at 50 μM concentration showed no toxicity on HUVECs, but attenuated LPS-induced injury, as evidenced by increased cell viability, reduction in LDH level and inflammatory cytokines in culture media. SAL also reduced MDA level and increased SOD activity in HUVECs, and inhibited apoptosis rate and caspase-3 activity. (P < 0.05). Moreover, LPS enhanced HUVECs autophagy, and SAL pretreatment further enhanced autophagy, with increased Beclin-1 protein and decreased P62 protein. SAL also attenuated LPS-induced activation of NLRP3 inflammasome, reduced the protein expression of NLRP3-related proteins, including ASC and caspase-1. Autophagy inhibition by 3-MA markedly reversed SAL-modulated changes in cell viability and NLRP3 expression in LPS-stimulated HUVECs. CONCLUSION SAL protects endothelial cells against LPS-induced injury through inhibition of NLRP3 pathways and enhancing autophagy.
Collapse
Affiliation(s)
- Lijiao You
- Department of Critical Care Medicine, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Gaoqiao Town, Pudong New District, Shanghai, 200137, China
| | - Di Zhang
- Department of Rehabilitation Medicine, Seventh People's Hospital of Shanghai University of TCM, Shanghai, 200137, P.R. China
| | - Huan Geng
- Department of Critical Care Medicine, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Gaoqiao Town, Pudong New District, Shanghai, 200137, China
| | - Fangyuan Sun
- Department of Critical Care Medicine, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Gaoqiao Town, Pudong New District, Shanghai, 200137, China
| | - Ming Lei
- Department of Critical Care Medicine, Seventh People's Hospital of Shanghai University of TCM, No.358 Datong Road, Gaoqiao Town, Pudong New District, Shanghai, 200137, China.
| |
Collapse
|
20
|
Hu S, Pi Q, Luo M, Cheng Z, Liang X, Luo S, Xia Y. Contribution of the NLRP3/IL-1β axis to impaired vasodilation in sepsis through facilitation of eNOS proteolysis and the protective role of melatonin. Int Immunopharmacol 2021; 93:107388. [PMID: 33529913 DOI: 10.1016/j.intimp.2021.107388] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/27/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
Endothelial dysfunction is a typical characteristic of sepsis. Endothelial nitric oxide synthase (eNOS) is important for maintaining endothelial function. Our previous study reported that the NLRP3 inflammasome promoted endothelial dysfunction by enhancing inflammation. However, the effects of NLRP3 on eNOS require further investigation. Therefore, the present study aimed to investigate the role of NLRP3 on eNOS expression levels in cecal ligation and puncture-induced impaired endothelium-dependent vascular relaxation and to determine the protective effects of melatonin. eNOS expression levels were discovered to be downregulated in the mesenteric arteries of sepsis model mice. Inhibiting NLRP3 with 10 mg/ kg MCC950 or inhibiting IL-1β with 100 mg diacerein rescued the eNOS expression and improved endothelium-dependent vascular relaxation. In vitro, IL-1β stimulation downregulated eNOS expression levels in human aortic endothelial cells (HAECs) in a concentration- and time-dependent manner, while pretreatment with 1 µM of the proteasome inhibitor MG132 reversed this effect. In addition, treatment with 10 mg/kg MG132 also prevented the proteolysis of eNOS and improved endothelium-dependent vascular relaxation in vivo. Notably, treatment with 30 mg/kg melatonin downregulated NLRP3 expression levels and decreased IL-1β secretion, subsequently increasing the expression of eNOS and improving endothelium-dependent vascular relaxation. In conclusion, the findings of the present study indicated that the NLRP3/IL-1β axis may impair vasodilation by promoting the proteolysis of eNOS and melatonin may protect against sepsis-induced endothelial relaxation dysfunction by inhibiting the NLRP3/IL-1β axis, suggesting its pharmacological potential in sepsis.
Collapse
Affiliation(s)
- Shupeng Hu
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Qiangzhong Pi
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Minghao Luo
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Zhe Cheng
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoxue Liang
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Suxin Luo
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Yong Xia
- Department of Cardiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China; Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, 473 West 12th Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
21
|
Lv D, Luo M, Yan J, Yang X, Luo S. Protective Effect of Sirtuin 3 on CLP-Induced Endothelial Dysfunction of Early Sepsis by Inhibiting NF-κB and NLRP3 Signaling Pathways. Inflammation 2021; 44:1782-1792. [PMID: 33770326 DOI: 10.1007/s10753-021-01454-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/07/2023]
Abstract
It has been revealed that widespread vascular endothelial dysfunction occurs in septic shock, ultimately resulting in multiple organ failure. The mitochondrial deacetylase sirtuin 3 (SIRT3) is essential in the regulation of metabolism, anti-inflammation, and anti-oxidation. The purpose of this study is to investigate whether SIRT3 is associated with the pathological progression of endothelial dysfunction in sepsis. Septic shock model was induced by cecal ligation and puncture (CLP) surgery on wild-type C57BL/6 mice. We activated and inhibited the function of SIRT3 with honokiol (HKL) and 3-TYP, respectively, and then biochemical, inflammatory, and endothelial function parameters of vascular tissue and survival were determined after CLP. CLP significantly activated NF-κB and NLRP3 pathways and decreased survival rate, endothelium-dependent relaxation function, and expression of Ser1177 phosphorylation of endothelial nitric oxide synthase (p-eNOS). The activation of SIRT3 significantly attenuated the increases of NF-κB and NLRP3 pathways and the declines of p-eNOS, endothelium-dependent relaxation function, and survival rate in septic mice. However, it presented exactly opposite results if SIRT3 was suppressed. We suggested that SIRT3 had a critical protective effect against vascular inflammation and endothelial dysfunction in early sepsis. Our data support a potential therapeutic target in vascular dysfunction and septic shock.
Collapse
Affiliation(s)
- Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Institute of Life Science, Chongqing Medical University, Chongqing, 400016, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Institute of Life Science, Chongqing Medical University, Chongqing, 400016, China
| | - Jianghong Yan
- Institute of Life Science, Chongqing Medical University, Chongqing, 400016, China
| | - Xiyang Yang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Institute of Life Science, Chongqing Medical University, Chongqing, 400016, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Institute of Life Science, Chongqing Medical University, Chongqing, 400016, China.
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
22
|
Chen ZB, Li G, Lin H, Jiang J, Jiang R. Low androgen status inhibits erectile function by increasing pyroptosis in rat corpus cavernosum. Andrology 2021; 9:1264-1274. [PMID: 33657666 DOI: 10.1111/andr.12995] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/01/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mechanism of erectile dysfunction (ED) caused by low androgen status is not fully understood. OBJECTIVES To investigate whether low androgen status inhibits erectile function of rats by inducing pyroptosis in the corpus cavernosum (CC). MATERIALS AND METHODS Thirty-six eight-weeks-old healthy male Sprague-Dawley rats were equally divided into six groups: sham-operated group (4w sham, 8w sham), castration group (4w cast, 8w cast), and castration + testosterone (T) group (4w cast + T, 8w cast + T). The rats in castration + T groups were injected with testosterone propionate subcutaneously every other day. After 4 and 8 weeks, the ratio of maximum intracavernous pressure (ICPmax)/mean arterial pressure (MAP), the level of serum T, the concentration of nitric oxide (NO) and interleukin-1β (IL-1β), the expression of NOD-like receptor pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), Caspase-1 p20, gasdermin D-N (GSDMD-N), transforming growth factor β1 (TGF-β1), collagen-I, and collagen-III, the ratio of smooth muscle/collagen (SM/C), and the proportion of pyroptotic cells in the CC were analyzed. RESULTS The ratio of ICPmax/MAP (3/5 V) and SM/C, the level of NO and serum T was significantly decreased in castration groups when compared to other groups (p < 0.01). NLRP3, ASC, Caspase-1, and GSDMD were mainly expressed in the cytoplasm of smooth muscle cells (SMCs) and endothelial cells (ECs) in the CC. The expression of NLRP3, ASC, Caspase-1p20, GSDMD-N, IL-1β, TGF-β1, collagen-I, and collagen-III was significantly increased in castration groups when compared with other groups (p < 0.01). The proportion of pyroptotic cells in the CC was increased significantly in castration groups when compared with other groups (p < 0.05). DISCUSSION AND CONCLUSION Low androgen status inhibits erectile function of rats by promoting CC fibrosis and reducing NO synthesis through pyroptosis of SMCs and ECs in the CC.
Collapse
Affiliation(s)
- Zhi-Bin Chen
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ge Li
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Haocheng Lin
- Department of Urology and Andrology, Peking University Third Hospital, Beijing, China
| | - Jun Jiang
- Department of Thyroid Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Rui Jiang
- Department of Urology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Nephropathy Clinical Medical Research Center of Sichuan Province, Luzhou, Sichuan, China
| |
Collapse
|
23
|
microRNA-103a-3p confers protection against lipopolysaccharide-induced sepsis and consequent multiple organ dysfunction syndrome by targeting HMGB1. INFECTION GENETICS AND EVOLUTION 2020; 89:104681. [PMID: 33333289 DOI: 10.1016/j.meegid.2020.104681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/08/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sepsis and subsequent multiple organ dysfunction syndrome (MODS) have high global incidence and mortality rate, imposing tremendous health burden. microRNAs (miRNAs or miRs) are implicated in the pathogenesis of sepsis and MODS. The aim of this study is to explore the potential mechanisms of miR-103a-3p targeted high mobility group box 1 (HMGB1) involvement in the pathogenesis of sepsis complicated with multiple organ dysfunction syndrome (MODS). METHODS A mouse sepsis model was induced by lipopolysaccharide (LPS). Bone marrow-derived macrophages were collected and LPS was used to establish a cellular inflammation model. Targeted binding between miR-103a-3p and HMGB1 was verified by a double luciferase assay and their roles in LPS-induced sepsis were further explored using gain-of-function experiments. RESULTS miR-103a-3p was decreased while HMGB1 was increased in sepsis. In LPS-induced mouse sepsis models, the downregulation of HMGB1 was found to result in reductions in NO, TNF-α, IL-1β, IL-6, lung myeloperoxidase activity, pulmonary microvascular albumin leakage, serum alanine aminotransferase, aspartate aminotransferase activity, and lung and liver tissue apoptosis. Additionally, decreased HMGB1 blunted the inflammatory response and increased survival rate of modeled mice. Importantly, HMGB1 was confirmed to a target gene of miR-103a-3p. In cellular inflammation models, miR-103a-3p was found to alleviate LPS-induced sepsis and MODS in vitro by decreasing HMGB1. CONCLUSIONS Taken together, our results demonstrated the inhibitory role of miR-103a-3p in sepsis via inhibiting HMGB1 expression.
Collapse
|
24
|
Bai B, Yang Y, Wang Q, Li M, Tian C, Liu Y, Aung LHH, Li PF, Yu T, Chu XM. NLRP3 inflammasome in endothelial dysfunction. Cell Death Dis 2020; 11:776. [PMID: 32948742 PMCID: PMC7501262 DOI: 10.1038/s41419-020-02985-x] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 12/24/2022]
Abstract
Inflammasomes are a class of cytosolic protein complexes. They act as cytosolic innate immune signal receptors to sense pathogens and initiate inflammatory responses under physiological and pathological conditions. The NLR-family pyrin domain-containing protein 3 (NLRP3) inflammasome is the most characteristic multimeric protein complex. Its activation triggers the cleavage of pro-interleukin (IL)-1β and pro-IL-18, which are mediated by caspase-1, and secretes mature forms of these mediators from cells to promote the further inflammatory process and oxidative stress. Simultaneously, cells undergo pro-inflammatory programmed cell death, termed pyroptosis. The danger signals for activating NLRP3 inflammasome are very extensive, especially reactive oxygen species (ROS), which act as an intermediate trigger to activate NLRP3 inflammasome, exacerbating subsequent inflammatory cascades and cell damage. Vascular endothelium at the site of inflammation is actively involved in the regulation of inflammation progression with important implications for cardiovascular homeostasis as a dynamically adaptable interface. Endothelial dysfunction is a hallmark and predictor for cardiovascular ailments or adverse cardiovascular events, such as coronary artery disease, diabetes mellitus, hypertension, and hypercholesterolemia. The loss of proper endothelial function may lead to tissue swelling, chronic inflammation, and the formation of thrombi. As such, elimination of endothelial cell inflammation or activation is of clinical relevance. In this review, we provided a comprehensive perspective on the pivotal role of NLRP3 inflammasome activation in aggravating oxidative stress and endothelial dysfunction and the possible underlying mechanisms. Furthermore, we highlighted the contribution of noncoding RNAs to NLRP3 inflammasome activation-associated endothelial dysfunction, and outlined potential clinical drugs targeting NLRP3 inflammasome involved in endothelial dysfunction. Collectively, this summary provides recent developments and perspectives on how NLRP3 inflammasome interferes with endothelial dysfunction and the potential research value of NLRP3 inflammasome as a potential mediator of endothelial dysfunction.
Collapse
Affiliation(s)
- Baochen Bai
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yanyan Yang
- Department of lmmunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Qi Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China
| | - Chao Tian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China
| | - Lynn Htet Htet Aung
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China.
- Department of Cardiac Ultrasound, The Affiliated hospital of Qingdao University, Qingdao, 266000, China.
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, 266032, China.
| |
Collapse
|
25
|
Anisodamine Hydrobromide Protects Glycocalyx and Against the Lipopolysaccharide-Induced Increases in Microvascular Endothelial Layer Permeability and Nitric Oxide Production. Cardiovasc Eng Technol 2020; 12:91-100. [PMID: 32935201 DOI: 10.1007/s13239-020-00486-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/08/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Anisodamine hydrobromide (Ani HBr) has been used to improve the microcirculation during cardiovascular disorders and sepsis. Glycocalyx plays an important role in preserving the endothelial cell (EC) barrier permeability and nitric oxide (NO) production. We aimed to test the hypothesis that Ani HBr could protect the EC against permeability and NO production via preventing glycocalyx shedding. METHODS A human cerebral microvascular EC hCMEC/D3 injury model induced by lipopolysaccharide (LPS) was established. Ani HBr was administrated to ECs with the LPS challenge. Cell viability was performed by Cell Counting Kit-8 assay. Cell proliferation and apoptosis were detected by EdU and Hoechst 33342 staining. Apoptosis and cell cycle were also assessed by flow cytometry with annexin V staining and propidium iodide staining, respectively. Then, adherens junction integrity was evaluated basing on the immunofluorescence staining of vascular endothelial cadherin (VE-cadherin). The glycocalyx component heparan sulfate (HS) was stained in ECs. The cell permeability was evaluated by leakage of fluorescein isothiocyanate (FITC)-dextran. Cellular NO production was measured by the method of nitric acid reductase. RESULTS Ani HBr at 20 μg/mL significantly increased the viability of ECs with LPS challenge, but significantly inhibited the cell viability at 80 μg/mL, showing a bidirectional regulation of cell viability by Ani HBr. Ani HBr had not significantly change the LPS-induced EC proliferation. Ani HBr significantly reversed the induction of LPS on EC apoptosis. Ani HBr reinstated the LPS-induced glycocalyx and VE-cadherin shedding and adherens junction disruption. Ani HBr significantly alleviated LPS-induced EC layer permeability and NO production. CONCLUSION Ani HBr protects ECs against LPS-induced increase in cell barrier permeability and nitric oxide production via preserving the integrity of glycocalyx. Ani HBr is a promising drug to rescue or protect the glycocalyx.
Collapse
|