51
|
Feng X, Chen Z, Cheng W, Liu C, Liu Q. Role for NLRP3 inflammasome-mediated, Caspase1-dependent response in glaucomatous trabecular meshwork cell death and regulation of aqueous humor outflow. Heliyon 2024; 10:e38258. [PMID: 39416828 PMCID: PMC11481635 DOI: 10.1016/j.heliyon.2024.e38258] [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: 01/02/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose Acute ocular hypertension (AOH) is the defining feature of acute glaucoma. The mechanical stress and excessive production of reactive oxygen species (ROS) during episodes can directly or indirectly damage the trabecular meshwork (TM). Despite its significance, a clear understanding of its pathogenesis and an effective therapeutic target remain lacking in acute glaucoma. In the present study, we explored the potential molecular mechanisms underlying TM cell death following oxidative damage and AOH. The use of NAC/VX-765 as a potential pharmaceutical intervention for reducing intraocular pressure (IOP) was discussed. Methods The levels of NLRP3 and caspase-1 were compared between normal and glaucomatous TM samples. An in vitro oxidative damage model and an AOH rat model were used to investigate the potential molecular mechanism behind TM cell death. The ROS scavenger N-acetyl-L-cysteine (NAC) and caspase-1 inhibitor VX-765 were used to counteract TM damage. Results Elevated levels of NLRP3 and caspase-1 were observed in patients with acute glaucoma. H2O2 exposure decreased the viability of human trabecular meshwork (HTM) cells and increased intracellular ROS levels. Both Gene and protein expressions of NLRP3, caspase-1, GSDMD-N, and IL-1β were notably upregulated in H2O2-induced HTM cells and the rodent AOH model. Both NAC and VX-765 demonstrated protective effects against TM injury by inhibiting pyroptosis. The IOP-lowering effects of NAC and VX-765 persisted for 7 days. Conclusions Our findings indicate that the classical pyroptosis pathway, NLRP3/caspase-1/IL-1β, plays a key role in acute glaucomatous TM injury. Targeting pyroptosis provides novel therapeutic avenues for treating AOH-induced irreversible TM injury. This provides not only a promising therapeutic target for glaucoma but also introduces a new approach to intervention.
Collapse
Affiliation(s)
- Xiaomei Feng
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, Zhengzhou, China
| | - Zhao Chen
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, Zhengzhou, China
| | - Wenjun Cheng
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, Zhengzhou, China
| | - Changgeng Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, Zhengzhou, China
| | - Qian Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan Eye Hospital, Henan Eye Institute, Zhengzhou, China
| |
Collapse
|
52
|
Luo J, Wu X, Chen D, Yu B, He J. Dietary ferulic acid supplementation enhances antioxidant capacity and alleviates hepatocyte pyroptosis in diquat challenged piglets. J Anim Sci Biotechnol 2024; 15:134. [PMID: 39370514 PMCID: PMC11457559 DOI: 10.1186/s40104-024-01086-5] [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: 05/23/2024] [Accepted: 08/04/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Oxidative stress significantly impacts growth performance and liver function in piglets. Ferulic acid (FA) works as an antioxidant, however, the role and mechanism of FA in the regulation of diquat-induced oxidative stress in piglets are less known. This study was designed to investigate the effects of FA on growth performance and antioxidant capacity in piglets with diquat challenge. METHODS Thirty-two healthy DLY (Duroc × Landrace × Yorkshire) piglets (13.24 ± 0.19 kg) were randomly divided into one of two diets including 0 or 4 g/kg FA for 14 d. On d 15, all pigs were intraperitoneally injected diquat or sterile saline. RESULTS Dietary supplementation with ferulic acid (FA) significantly improved the average daily gain (ADG) and decreased feed-gain ratio (F/G) of piglets. Here, dietary FA supplementation reduced serum aspartate aminotransferase (AST), alanine aminotransferase (ALT) activities in diquat challenged piglets. Furthermore, diquat infusion increased reactive oxygen radicals (ROS) level in liver, decreased the activities of total superoxide dismutase (T-SOD) and glutathione peroxidase (GSH-Px), total antioxidant capacity (T-AOC) and increased malondialdehyde (MDA) content in the liver and serum. Supplementation with FA significantly increased T-AOC and T-SOD activities and decreased MDA and ROS levels. FA down-regulated gene and protein expression of Keap1, and up-regulated protein expression of Nrf2 and HO-1 in the liver of piglets with diquat challenge. Importantly, diquat challenge increased the ratio of late apoptosis, increased serum levels of IL-1β, IL-18 and lactate dehydrogenase (LDH), and up-regulated pyroptosis-related genes in the liver. FA supplementation reduced the ratio of late apoptosis and down-regulated mRNA expression of Caspase-1. Accordingly, FA addition reduced concentration of IL-1β, IL-18, and LDH under diquat challenge. CONCLUSIONS Diquat-induced oxidative stress reduced growth performance and impaired liver function in piglets. Dietary FA supplementation enhanced the antioxidant capacity and reduced the degree of hepatocyte pyroptosis, thereby alleviating the oxidative damage in the liver and mitigating the impact of diquat on growth performance of piglets.
Collapse
Affiliation(s)
- Junqiu Luo
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.
| | - Xiu Wu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Daiwen Chen
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China
| |
Collapse
|
53
|
Liu L, Shi J, Wang J, He L, Gao Y, Lin P, Han Y, Ma P, Lin J, Zhang Y. Biodegradable Persistent Luminescence Nanoparticles as Pyroptosis Inducer for High-Efficiency Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406340. [PMID: 39158490 PMCID: PMC11497027 DOI: 10.1002/advs.202406340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/05/2024] [Indexed: 08/20/2024]
Abstract
Pyroptosis possesses potent antitumor immune activity, making pyroptosis inducer development a promising direction for tumor immunotherapy. Persistent luminescence nanoparticles (PLNPs) are highly sensitive optical probes extensively employed in tumor diagnosis and therapy. However, a pyroptosis inducer based on PLNPs has not been reported yet. Herein, polyethylene glycol-poly lactic acid-co-glycolic acid (PEG-PLGA: PP) modified biodegradable CaS:Eu2+ (CSE@PP) PLNPs are synthesized as a pyroptosis inducer for tumor immunotherapy for the first time. The synthesized CSE@PP possesses biowindow persistent luminescence (PersL) and pH-responsive degradation properties, allowing it to remain stable under neutral pH but degrade when exposed to weak acid (pH < 6.5). During degradation within the tumor, CSE@PP constantly releases H2S and Ca2+ while its PersL gradually fades away. Thus, the PersL signal can self-monitor H2S and Ca2+ release. Furthermore, the released H2S and Ca2+ result in mitochondrial dysfunction and the inactivation of reactive oxygen species scavenging enzymes, synergistic facilitating intracellular oxidative stress, which induces caspase-1/GSDM-D dependent pyroptosis and subsequent antitumor immune responses. In a word, it is confirmed that CSE@PP can self-monitor H2S and Ca2+ release and pyroptosis-mediated tumor Immunotherapy. This work will facilitate biomedical applications of PLNPs and inspire pyroptosis-induced tumor immunotherapy.
Collapse
Affiliation(s)
- Lin Liu
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamen361021China
- University of Chinese Academy of SciencesBeijing100049China
| | - Junpeng Shi
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamen361021China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jinyuan Wang
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamen361021China
| | - Linping He
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamen361021China
| | - Yan Gao
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamen361021China
| | - Peng Lin
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamen361021China
| | - Yutong Han
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt LuciaQLD4067Australia
| | - Ping'an Ma
- University of Chinese Academy of SciencesBeijing100049China
- State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022China
| | - Jun Lin
- University of Chinese Academy of SciencesBeijing100049China
- State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022China
| | - Yun Zhang
- State Key Laboratory of Structural ChemistryFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional MaterialsXiamen Institute of Rare Earth MaterialsHaixi InstituteChinese Academy of SciencesXiamen361021China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
54
|
Lin X, Zhuang Y, Gao F. ACE2 Alleviates Endoplasmic Reticulum Stress and Protects against Pyroptosis by Regulating Ang1-7/Mas in Ventilator-Induced Lung Injury. FRONT BIOSCI-LANDMRK 2024; 29:334. [PMID: 39344337 DOI: 10.31083/j.fbl2909334] [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: 11/06/2023] [Revised: 12/19/2023] [Accepted: 02/04/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Ventilator-induced lung injury (VILI) is a consequence of inflammation and increased alveolar-capillary membrane permeability due to alveolar hyperdistention or elevated intrapulmonary pressure, but the precise mechanisms remain unclear. The aim of the study was to analyze the mechanism by which angiotensin converting enzyme 2 (ACE2) alleviates endoplasmic reticulum stress (ERS) and protects alveolar cells from pyroptosis in VILI by regulating angiotensin (Ang)1-7/Mas. METHODS VILI was induced in mice by mechanical ventilation by regulating the tidal volume. The alveolar cell line, A549, mimics VILI in vitro by cyclic stretch (CS). Ang (1-7) (100 nmol/L) was added to the medium. ERS was induced in cells by stimulating with tunicamycin (TM, 2 μg/mL). ERS was inhibited by tracheal instillation of 4-phenylbutyric acid (4-PBA) (1 mg/kg). ACE2's enzymatic function was activated or inhibited by subcutaneous injection of resorcinolnaphthalein (RES, 20 μg/kg) or MLN-4760 (20 μg/kg). pGLV-EF1a-GFP-ACE2 was instilled into the trachea to increase the protein expression of ACE2. The Ang (1-7) receptor, Mas, was antagonized by injecting A779 subcutaneously (80 μg/kg). RESULTS ACE2 protein levels decreased after modeling. Ang (1-7) level was decreased and Ang II was accumulated. ERS was significantly induced in VILI mice, and pyroptosis was observed in cells. When ERS was inhibited, pyroptosis under the VILI condition was significantly inhibited. Ang (1-7) alleviated ERS and pyroptosis under CS. When ERS was continuously activated, the function of Ang (1-7) in inhibiting pyroptosis was blocked. Resorcinolnaphthalein (RES) effectively promoted Ang II conversion, alleviated the Ang (1-7) level in VILI, ameliorated lung injury, and inhibited ERS and cell pyroptosis. Inhibiting ACE2's function in VILI hindered the production of Ang (1-7), promoted the accumulation of Ang II, and exacerbated ERS and pyroptosis, along with lung injury. The Mas antagonist significantly blocked the inhibitory effects of ACE2 on ERS and pyroptosis in VILI. CONCLUSIONS Reduced ACE2 expression in VILI is involved in ERS and pyroptosis-related injury. ACE2 can alleviate ERS in alveolar cells by catalyzing the production of Ang (1-7), thus inhibiting pyroptosis in VILI.
Collapse
Affiliation(s)
- Xingsheng Lin
- Department of Intensive Care Unit, Fuzhou University Affiliated Provincial Hospital, 350001 Fuzhou, Fujian, China
| | - Yingfeng Zhuang
- Department of Intensive Care Unit, Fuzhou University Affiliated Provincial Hospital, 350001 Fuzhou, Fujian, China
| | - Fengying Gao
- Department of Pulmonary Disease, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071 Shanghai, China
| |
Collapse
|
55
|
Wang K, Xie X, Hu X, Wang Z, Xia J, Wu Q. Stearic acid alleviates aortic medial degeneration through maintaining mitochondrial dynamics homeostasis via inhibiting JNK/MAPK signaling. iScience 2024; 27:110594. [PMID: 39224510 PMCID: PMC11367538 DOI: 10.1016/j.isci.2024.110594] [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: 03/20/2024] [Revised: 06/14/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Aortic dissection is characterized pathologically by aortic medial degeneration (AMD) where disturbance of mitochondrial dynamics may be involved. Stearic acid (SA) can promote mitochondrial fusion and improve mitochondrial function. Here, we established an AMD mouse model through oral administration of β-aminopropionitrile (BAPN) and a cellular model by treating primary vascular smooth muscle cells (VSMCs) with Angiotensin-II to explore the potential role of SA in AMD. Our results showed SA reduced AMD and prolonged survival of BAPN-treated mice. Excessive mitochondrial fission was observed during AMD both in vivo and in vitro, and SA reduced mitochondrial fission and increased fusion. Additionally, SA promoted expression of contractile phenotype markers of VSMCs. At the molecular level, SA reduced AMD by inhibiting JNK/MAPK signaling. Our study suggests SA can promote mitochondrial fusion and increase the contractile phenotype of VSMCs by inhibiting JNK/MAPK signaling, thereby reducing AMD formation and possibly the consequent risk of aortic dissection.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Central Laboratory, Renmin Hospital of Wuhan University, 9 Zhangzhidong Road, Wuhan 430060, Hubei Province, P.R. China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
| | - Xiaoping Xie
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Central Laboratory, Renmin Hospital of Wuhan University, 9 Zhangzhidong Road, Wuhan 430060, Hubei Province, P.R. China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
| | - Xiaoping Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Central Laboratory, Renmin Hospital of Wuhan University, 9 Zhangzhidong Road, Wuhan 430060, Hubei Province, P.R. China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Central Laboratory, Renmin Hospital of Wuhan University, 9 Zhangzhidong Road, Wuhan 430060, Hubei Province, P.R. China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
| | - Jun Xia
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Central Laboratory, Renmin Hospital of Wuhan University, 9 Zhangzhidong Road, Wuhan 430060, Hubei Province, P.R. China
| | - Qi Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, Hubei Province, P.R. China
- Central Laboratory, Renmin Hospital of Wuhan University, 9 Zhangzhidong Road, Wuhan 430060, Hubei Province, P.R. China
| |
Collapse
|
56
|
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
|
57
|
Long H, Yu Y, Ouyang J, Lu H, Zhao G. Insights into RNA N6-methyladenosine and programmed cell death in atherosclerosis. Mol Med 2024; 30:137. [PMID: 39227813 PMCID: PMC11373444 DOI: 10.1186/s10020-024-00901-z] [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: 07/07/2024] [Accepted: 08/18/2024] [Indexed: 09/05/2024] Open
Abstract
N6-methyladenosine (m6A) modification stands out among various RNA modifications as the predominant form within eukaryotic cells, influencing numerous cellular processes implicated in disease development. m6A modification has gained increasing attention in the development of atherosclerosis and has become a research hotspot in recent years. Programmed cell death (PCD), encompassing apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis, plays a pivotal role in atherosclerosis pathogenesis. In this review, we delve into the intricate interplay between m6A modification and diverse PCD pathways, shedding light on their complex association during the onset and progression of atherosclerosis. Clarifying the relationship between m6A and PCD in atherosclerosis is of great significance to provide novel strategies for cardiovascular disease treatment.
Collapse
Affiliation(s)
- Haijiao Long
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Yulu Yu
- Afliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, Guangdong, China
| | - Jie Ouyang
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Hongwei Lu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Guojun Zhao
- Afliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, Guangdong, China.
| |
Collapse
|
58
|
He H, Chen J, Hua Y, Xie Z, Tu M, Liu L, Wang H, Yang X, Chen L. α7-nAChR/P300/NLRP3-regulated pyroptosis mediated poor articular cartilage quality induced by prenatal nicotine exposure in female offspring rats. Chem Biol Interact 2024; 400:111183. [PMID: 39098741 DOI: 10.1016/j.cbi.2024.111183] [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: 05/20/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Nicotine is developmentally toxic. Prenatal nicotine exposure (PNE) affects the development of multiple fetal organs and causes susceptibility to a variety of diseases in offspring. In this study, we aimed to investigate the effect of PNE on cartilage development and osteoarthritis susceptibility in female offspring rats. Wistar rats were orally gavaged with nicotine on days 9-20 of pregnancy. The articular cartilage was obtained at gestational day (GD) 20 and postnatal week (PW) 24, respectively. Further, the effect of nicotine on chondrogenic differentiation was explored by the chondrogenic differentiation model in human Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs). The PNE group showed significantly shallower Safranin O staining and lower Collagen 2a1 content of articular cartilage in female offspring rats. Further, we found that PNE activated pyroptosis in the articular cartilage at GD20 and PW24. In vitro experiments revealed that nicotine inhibited chondrogenic differentiation and activated pyroptosis. After interfering with nod-like receptors3 (NLRP3) expression by SiRNA, it was found that pyroptosis mediated the chondrogenic differentiation inhibition of WJ-MSCs induced by nicotine. In addition, we found that α7-nAChR antagonist α-BTX reversed nicotine-induced NLRP3 and P300 high expression. And, P300 SiRNA reversed the increase of NLRP3 mRNA expression and histone acetylation level in its promoter region induced by nicotine. In conclusion, PNE caused chondrodysplasia and poor articular cartilage quality in female offspring rats. PNE increased the histone acetylation level of NLRP3 promoter region by α7-nAChR/P300, which resulting in the high expression of NLRP3. Further, NLRP3 mediated the inhibition of chondrogenic differentiation by activating pyroptosis.
Collapse
Affiliation(s)
- Hangyuan He
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jun Chen
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yi Hua
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhe Xie
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Tu
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Liang Liu
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Xu Yang
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Liaobin Chen
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
59
|
Li Q, Pang B, Dang E, Wang G. Endothelial Dysfunction in Psoriasis: An Integrative Review. J Invest Dermatol 2024; 144:1935-1942. [PMID: 38493385 DOI: 10.1016/j.jid.2024.02.013] [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: 10/30/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 03/18/2024]
Abstract
Vascular endothelial cells (ECs), the inner layer of blood vessels, were previously considered to be a passive lining that facilitates cellular and molecular exchange. However, recent studies have revealed that ECs can respond to various stimuli and actively regulate vascular function and skin inflammation. Specific subtypes of ECs are known to have significant roles in a diverse range of physiological and pathological processes in the skin. This review suggests that EC dysfunction is both causal and consequential in the pathogenesis of psoriasis. Further investigations into dysregulated pathways in EC dysfunction may provide new insights for the treatment of psoriasis.
Collapse
Affiliation(s)
- Qingyang Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People Republic of China
| | - Bingyu Pang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People Republic of China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People Republic of China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People Republic of China.
| |
Collapse
|
60
|
Zhang W, Li Y, Zheng K, Li Y, Yang H. Nonlinear associations between dietary zinc intake and cardiovascular disease risk, a National cross-sectional study based on the NHANES 2005-2018. Prev Med Rep 2024; 45:102830. [PMID: 39132580 PMCID: PMC11315215 DOI: 10.1016/j.pmedr.2024.102830] [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: 01/11/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 08/13/2024] Open
Abstract
Objective To explore the associations between dietary zinc intake and cardiovascular diseases (CVDs), including congestive heart failure (CHF), coronary heart disease (CHD), angina, heart attack, and cerebrovascular accident (CVA), this study was performed. Setting Data from the National Health and Nutrition Examination Survey (2005-2018) were used in this study. Dietary zinc intake was stratified into quartiles. Restricted cubic splines were constructed to assess nonlinear associations and identify cut-off values based on the type of nonlinearity. Binary logistic regressions were performed using the cut-offs. Results Positive associations were detected between the second, third, and fourth quantiles of dietary zinc intake and decreased risks of overall CVDs (Q2: OR = 0.83, 95 % CI = 0.72-0.96; Q3: OR = 0.83, 95 % CI = 0.71-0.96; Q4: OR = 0.79, 95 % CI = 0.67-0.93). The second, third, and fourth quantiles were significantly associated with decreased risks of various CVDs (all P < 0.05), except for CHD and angina (all P > 0.05). Restricted cubic spline regression revealed significant nonlinear trends for associations of dietary zinc intake with the risk of developing CVDs and CHF (both P for nonlinear <0.05), whereas those for heart attack and CVA were marginally significant (P for nonlinear = 0.072, and 0.075, respectively). Conclusions This study revealed that high dietary zinc intake is associated with reduced risks of developing CVDs, CHF, heart attack, and CVA, but not CHD or angina.
Collapse
Affiliation(s)
- Weihao Zhang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guizhou 610041, China
| | - Yuming Li
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guizhou 610041, China
| | - Kai Zheng
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guizhou 610041, China
| | - Yuanjing Li
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guizhou 610041, China
| | - Hua Yang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guizhou 610041, China
| |
Collapse
|
61
|
Wang X, Dong Y, Du H, Lu Y, Jiang Y, Ding M, Sheng X. Vascular endothelial cells of Mongolian gerbils are resistant to cholesterol-induced mitochondrial dysfunction and oxidative damage. Exp Ther Med 2024; 28:356. [PMID: 39071902 PMCID: PMC11273357 DOI: 10.3892/etm.2024.12645] [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: 01/06/2024] [Accepted: 06/19/2024] [Indexed: 07/30/2024] Open
Abstract
Atherosclerosis is essentially the leading factor behind occurrences of cardiovascular diseases (CVDs)-associated incidents, while mitochondrial dysfunction is also the main cause of atherosclerosis. The present study conducted a comparative analysis of mitochondrial function-related indicators in cholesterol-induced vascular endothelial cells (VECs) from Mongolian gerbils, Sprague-Dawley (SD) rats and humans. It reported that the inhibitory effect of cholesterol treatment on the viability of Mongolian gerbil VECs was markedly lower than the other two types of VECs at the same concentration. Following cholesterol treatment, mitochondrial DNA copy numbers, reactive oxygen species level, calcium concentration and mitochondrial membrane potential of Mongolian gerbil VECs did not change markedly. These results suggested that the function of mitochondria in the VECs of Mongolian gerbil is normal. Additionally, cholesterol treatment also did not alter the levels of superoxide dismutase, glutathione peroxidase, ATP, NADH-CoQ reductase and cytochrome c oxidase in Mongolian gerbil VECs. It was hypothesized that the VECs of Mongolian gerbils have certain resistance to oxidative damage induced by cholesterol. In brief, the present study demonstrated that VECs of Mongolian gerbils are resistant to cholesterol-induced mitochondrial dysfunction and oxidative damage. The aforementioned findings establish a theoretical foundation for the advancement of innovative strategies in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiaobing Wang
- School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang 321007, P.R. China
| | - Yuchen Dong
- School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang 321007, P.R. China
| | - Hongjian Du
- School of Pharmacy, Jinhua Polytechnic, Jinhua, Zhejiang 321007, P.R. China
| | - Yijia Lu
- School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang 321007, P.R. China
| | - Yanjie Jiang
- Institute of Pharmacology, Jinhua Food Institute for Food and Drug Control, Jinhua, Zhejiang 321000, P.R. China
| | - Mingxing Ding
- School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang 321007, P.R. China
| | - Xiaosheng Sheng
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
62
|
Cheng Q, Zhang T, Wang Q, Wu X, Li L, Lin R, Zhou Y, Qu S. Photocatalytic Carbon Dots-Triggered Pyroptosis for Whole Cancer Cell Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408685. [PMID: 39129656 DOI: 10.1002/adma.202408685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/25/2024] [Indexed: 08/13/2024]
Abstract
Manufacturing whole cancer cell vaccines (WCCV) with both biosafety and efficacy is crucial for tumor immunotherapy. Pyroptotic cancer cells, due to their highly immunogenic properties, present a promising avenue for the development of WCCV. However, the successful development of WCCV based on pyroptotic cancer cells is yet to be accomplished. Here, a facile strategy that utilized photocatalytic carbon dots (CDs) to induce pyroptosis of cancer cells for fabricating WCCV is reported. Photocatalytic CDs are capable of generating substantial amounts of hydroxyl radicals and can effectively decrease cytoplasmic pH values under white light irradiation. This process efficiently triggers cancer cell pyroptosis through the reactive oxygen species (ROS)-mitochondria-caspase 3-gasdermin E pathway and the proton motive force-driven mitochondrial ATP synthesis pathway. Moreover, in vitro, these photocatalytic CDs-induced pyroptotic cancer cells (PCIP) can hyperactivate macrophage (M0-M1) with upregulation of major histocompatibility complex class II expression. In vivo, PCIP induced specific immune-preventive effects in melanoma and breast cancer mouse models through anticancer immune memory, demonstrating effective WCCV. This work provides novel insights for inducing cancer cell pyroptosis and bridges the gap in the fabrication of WCCV based on pyroptotic cancer cells.
Collapse
Affiliation(s)
- Quansheng Cheng
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Tesen Zhang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Qingcheng Wang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Xue Wu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Lingyun Li
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Runxing Lin
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
- Department of Physics and Chemistry, Faculty of Science and Technology, University of Macau, Taipa, Macau, SAR, 999078, China
- MOE Frontier Science Centre for Precision Oncology, Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau, SAR, 999078, China
| | - Songnan Qu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau, SAR, 999078, China
- Department of Physics and Chemistry, Faculty of Science and Technology, University of Macau, Taipa, Macau, SAR, 999078, China
- MOE Frontier Science Centre for Precision Oncology, Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau, SAR, 999078, China
- Zhuhai UM Science and Technology Research Institute, Zhuhai, 519031, China
| |
Collapse
|
63
|
Gao X, Ma C, Liang S, Chen M, He Y, Lei W. PANoptosis: Novel insight into regulated cell death and its potential role in cardiovascular diseases (Review). Int J Mol Med 2024; 54:74. [PMID: 38963054 PMCID: PMC11254103 DOI: 10.3892/ijmm.2024.5398] [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: 01/25/2024] [Accepted: 05/08/2024] [Indexed: 07/05/2024] Open
Abstract
PANoptosis, a complex form of proinflammatory programmed cell death, including apoptosis, pyroptosis and necroptosis, has been an emerging concept in recent years that has been widely reported in cancer, infectious diseases and neurological disorders. Cardiovascular diseases (CVDs) are an important global health problem, posing a serious threat to individuals' lives. An increasing body of research shows that inflammation has a pivotal role in CVDs, which provides an important theoretical basis for PANoptosis to promote the progression of CVDs. To date, only sporadic studies on PANoptosis in CVDs have been reported and its role in the field of CVDs has not been fully explored. Elucidating the various modes of cardiomyocyte death, the specific molecular mechanisms and the links among the various modes of death under various stressful stimuli is of notable clinical significance for a deeper understanding of the pathophysiology of CVDs. The present review summarizes the molecular mechanisms of apoptosis, pyroptosis, necroptosis and PANoptosis and their prospects in the field of CVDs.
Collapse
Affiliation(s)
- Xinyu Gao
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Cuixue Ma
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Shan Liang
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Meihong Chen
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
64
|
Kiełbowski K, Skórka P, Plewa P, Bakinowska E, Pawlik A. The Role of Alarmins in the Pathogenesis of Atherosclerosis and Myocardial Infarction. Curr Issues Mol Biol 2024; 46:8995-9015. [PMID: 39194749 DOI: 10.3390/cimb46080532] [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: 06/29/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Atherosclerosis is a condition that is associated with lipid accumulation in the arterial intima. Consequently, the enlarging lesion, which is also known as an atherosclerotic plaque, may close the blood vessel lumen, thus leading to organ ischaemia. Furthermore, the plaque may rupture and initiate the formation of a thrombus, which can cause acute ischaemia. Atherosclerosis is a background pathological condition that can eventually lead to major cardiovascular diseases such as acute coronary syndrome or ischaemic stroke. The disorder is associated with an altered profile of alarmins, stress response molecules that are secreted due to cell injury or death and that induce inflammatory responses. High-mobility group box 1 (HMGB1), S100 proteins, interleukin-33, and heat shock proteins (HSPs) also affect the behaviour of endothelial cells and vascular smooth muscle cells (VSMCs). Thus, alarmins control the inflammatory responses of endothelial cells and proliferation of VSMCs, two important processes implicated in the pathogenesis of atherosclerosis. In this review, we will discuss the role of alarmins in the pathophysiology of atherosclerosis and myocardial infarction.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Patryk Skórka
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
65
|
Cao Y, Sun J, Wang X, Zhang X, Tian H, Huang L, Huang Z, Zhang Y, Zhang J, Li L, Zhou S. The double-edged nature of nicotine: toxicities and therapeutic potentials. Front Pharmacol 2024; 15:1427314. [PMID: 39206262 PMCID: PMC11350241 DOI: 10.3389/fphar.2024.1427314] [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: 05/03/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Nicotine is the primary addictive component of cigarette smoke and is associated with various smoking-related diseases. However, recent research has revealed its broader cognitive-enhancing and anti-inflammatory properties, suggesting its potential therapeutic applications in several conditions. This review aims to examine the double-edged nature of nicotine, encompassing its positive and negative effects. We provide a concise overview of the physiochemical properties and pharmacology of nicotine, including insights into nicotine receptors. Therefore, the article is divided into two main sections: toxicity and therapeutic potential. We comprehensively explored nicotine-related diseases, focusing on specific signaling pathways and the underlying mechanisms that contribute to its effects. Furthermore, we addressed the current research challenges and future development perspectives. This review aims to inspire future researchers to explore the full medical potential of nicotine, which holds significant promise for the clinical management of specific diseases.
Collapse
Affiliation(s)
- Yun Cao
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Jiali Sun
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, China
| | - Xiaofeng Wang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Xiaoyu Zhang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Huijuan Tian
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Lingling Huang
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Ze Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, China
| | - Yaping Zhang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
- Key Laboratory of Aerosol Analysis Regulation and Biological Effects of Anhui Province, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Jin Zhang
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, China
| | - Shun Zhou
- Key Laboratory of Combustion & Pyrolysis Study of CNTC, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
- Key Laboratory of Aerosol Analysis Regulation and Biological Effects of Anhui Province, China Tobacco Anhui Industrial Co., Ltd., Hefei, China
| |
Collapse
|
66
|
Bhat OM, Mir RA, Nehvi IB, Wani NA, Dar AH, Zargar MA. Emerging role of sphingolipids and extracellular vesicles in development and therapeutics of cardiovascular diseases. IJC HEART & VASCULATURE 2024; 53:101469. [PMID: 39139609 PMCID: PMC11320467 DOI: 10.1016/j.ijcha.2024.101469] [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: 03/18/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024]
Abstract
Sphingolipids are eighteen carbon alcohol lipids synthesized from non-sphingolipid precursors in the endoplasmic reticulum (ER). The sphingolipids serve as precursors for a vast range of moieties found in our cells that play a critical role in various cellular processes, including cell division, senescence, migration, differentiation, apoptosis, pyroptosis, autophagy, nutrition intake, metabolism, and protein synthesis. In CVDs, different subclasses of sphingolipids and other derived molecules such as sphingomyelin (SM), ceramides (CERs), and sphingosine-1-phosphate (S1P) are directly related to diabetic cardiomyopathy, dilated cardiomyopathy, myocarditis, ischemic heart disease (IHD), hypertension, and atherogenesis. Several genome-wide association studies showed an association between genetic variations in sphingolipid pathway genes and the risk of CVDs. The sphingolipid pathway plays an important role in the biogenesis and secretion of exosomes. Small extracellular vesicles (sEVs)/ exosomes have recently been found as possible indicators for the onset of CVDs, linking various cellular signaling pathways that contribute to the disease progression. Important features of EVs like biocompatibility, and crossing of biological barriers can improve the pharmacokinetics of drugs and will be exploited to develop next-generation drug delivery systems. In this review, we have comprehensively discussed the role of sphingolipids, and sphingolipid metabolites in the development of CVDs. In addition, concise deliberations were laid to discuss the role of sEVs/exosomes in regulating the pathophysiological processes of CVDs and the exosomes as therapeutic targets.
Collapse
Affiliation(s)
- Owais Mohmad Bhat
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Rakeeb Ahmad Mir
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | | | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Abid Hamid Dar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - M Afzal Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| |
Collapse
|
67
|
Scharf P, Sandri S, Borges PP, Franco de Oliveira T, Farsky SHP. A single and short exposure to heated tobacco vapor or cigarette smoke affects macrophage activation and polarization. Toxicology 2024; 506:153859. [PMID: 38825031 DOI: 10.1016/j.tox.2024.153859] [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: 04/04/2024] [Revised: 05/18/2024] [Accepted: 05/31/2024] [Indexed: 06/04/2024]
Abstract
The toxicity of heated tobacco products (HTP) on the immune cells remains unclear. Here, U937-differentiated macrophages were exposed to a single and short-term exposure (30 minutes) of HTP vapor or cigarette smoke (CS) in an air-liquid interface (ALI) system to evaluate the effects on macrophages' early activation and polarization. In our system, HTP released lower amounts of polycyclic aromatic hydrocarbons (PAHs), but higher nicotine levels than CS into the cell culture supernatant. Both tobacco products triggered the expression of the α-7 nicotinic receptor (α7 nAChR) and reactive oxygen species (ROS) production. When challenged with a bacterial product, lipopolysaccharide (LPS), cells exposed to HTP or CS failed to respond properly and enhance ROS production upon LPS stimuli. Furthermore, both tobacco products also impaired bacterial phagocytosis and the exposures triggered higher IL-1β secretion. The α7 nAChR antagonist treatment rescued the effects caused only by HTP exposure. The CS-exposed group switched macrophage to the pro-inflammatory M1, while HTP polarized to the suppressive M2 profile. Associated, data highlight that HTP and CS exposures similarly activate macrophages; nonetheless, the α7 nAChR pathway is only involved in HTP actions, and the distinct subsequent polarization caused by HTP or CS may influence the outcome of host defense.
Collapse
Affiliation(s)
- Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, SP 05508-000, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, SP 05508-000, Brazil
| | - Pâmela Pacassa Borges
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, SP 05508-000, Brazil
| | - Tiago Franco de Oliveira
- Department of Pharmacosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
68
|
Lei S, Liu C, Zheng TX, Fu W, Huang MZ. The relationship of redox signaling with the risk for atherosclerosis. Front Pharmacol 2024; 15:1430293. [PMID: 39148537 PMCID: PMC11324460 DOI: 10.3389/fphar.2024.1430293] [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: 05/09/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
Oxidative balance plays a pivotal role in physiological homeostasis, and many diseases, particularly age-related conditions, are closely associated with oxidative imbalance. While the strategic role of oxidative regulation in various diseases is well-established, the specific involvement of oxidative stress in atherosclerosis remains elusive. Atherosclerosis is a chronic inflammatory disorder characterized by plaque formation within the arteries. Alterations in the oxidative status of vascular tissues are linked to the onset, progression, and outcome of atherosclerosis. This review examines the role of redox signaling in atherosclerosis, including its impact on risk factors such as dyslipidemia, hyperglycemia, inflammation, and unhealthy lifestyle, along with dysregulation, vascular homeostasis, immune system interaction, and therapeutic considerations. Understanding redox signal transduction and the regulation of redox signaling will offer valuable insights into the pathogenesis of atherosclerosis and guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sujuan Lei
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Tian-Xiang Zheng
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Mei-Zhou Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| |
Collapse
|
69
|
Su YQ, Lin Y, Huang SJ, Lin YT, Ran J, Yan FF, Liu XL, Hong LC, Huang M, Su HZ, Zhang XD, You JH, Su YM. Pyroptosis is involved in maternal nicotine exposure-induced metabolic associated fatty liver disease progression in offspring mice. Mol Reprod Dev 2024; 91:e23719. [PMID: 38018308 DOI: 10.1002/mrd.23719] [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: 01/07/2023] [Revised: 10/29/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
We have investigated whether inflammasomes and pyroptosis are activated in maternal nicotine exposure (MNE) offspring mice and whether they are involved in MNE-promoted metabolic associated fatty liver disease (MAFLD) in adult offspring. We injected pregnant mice subcutaneously with saline vehicle or nicotine twice a day on gestational days 11-21. Offspring mice from both groups were fed with a normal diet (ND) or a high-fat diet (HFD) for 6 months at postnatal day 21 to develop the MAFLD model. Serum biochemical indices were analyzed, and liver histology was performed. The expression levels of inflammasome and pyroptosis proteins were detected by western blot. We found MNE significantly aggravated the injury of MAFLD in adult offspring mice. MNE activated inflammasomes and pyroptosis in both infant and adult offspring mice. HFD treatment activated inflammasomes but not pyroptosis at 3 months, while it showed no effect at 6 months. However, pyroptosis was more severe in MNE-HFD mice than in MNE-ND mice at 6 months. Taken together, our data suggest MNE promotes MAFLD progression in adult offspring mice. MNE also induces NLRP3 and NLRP6 inflammasome activation and pyroptosis in both infant and adult offspring mice, which may be involved in MNE-promoted progression of MAFLD.
Collapse
Affiliation(s)
- Yu-Qing Su
- Department of Ultrasound, The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yan Lin
- Department of Ultrasound, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Shu-Jing Huang
- Department of Ultrasound, The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yan-Ting Lin
- Department of Ultrasound, The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jing Ran
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Fang-Fang Yan
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xian-Lan Liu
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Long-Cheng Hong
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Mei Huang
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Huan-Zhong Su
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao-Dong Zhang
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jian-Hong You
- Department of Ultrasound, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yi-Ming Su
- Department of Ultrasound, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Ultrasound, Siming Branch Hospital, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Collaborative Innovation Center for Maternal and Infant Health Service Application technology, Quanzhou Medical College, Quanzhou, China
| |
Collapse
|
70
|
Li F, Sun X, Sun K, Kong F, Jiang X, Kong Q. Lupenone improves motor dysfunction in spinal cord injury mice through inhibiting the inflammasome activation and pyroptosis in microglia via the nuclear factor kappa B pathway. Neural Regen Res 2024; 19:1802-1811. [PMID: 38103247 PMCID: PMC10960275 DOI: 10.4103/1673-5374.389302] [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: 09/22/2022] [Revised: 07/27/2023] [Accepted: 09/13/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00034/figure1/v/2023-12-16T180322Z/r/image-tiff Spinal cord injury-induced motor dysfunction is associated with neuroinflammation. Studies have shown that the triterpenoid lupenone, a natural product found in various plants, has a remarkable anti-inflammatory effect in the context of chronic inflammation. However, the effects of lupenone on acute inflammation induced by spinal cord injury remain unknown. In this study, we established an impact-induced mouse model of spinal cord injury, and then treated the injured mice with lupenone (8 mg/kg, twice a day) by intraperitoneal injection. We also treated BV2 cells with lipopolysaccharide and adenosine 5'-triphosphate to simulate the inflammatory response after spinal cord injury. Our results showed that lupenone reduced IκBα activation and p65 nuclear translocation, inhibited NLRP3 inflammasome function by modulating nuclear factor kappa B, and enhanced the conversion of proinflammatory M1 microglial cells into anti-inflammatory M2 microglial cells. Furthermore, lupenone decreased NLRP3 inflammasome activation, NLRP3-induced microglial cell polarization, and microglia pyroptosis by inhibiting the nuclear factor kappa B pathway. These findings suggest that lupenone protects against spinal cord injury by inhibiting inflammasomes.
Collapse
Affiliation(s)
- Fudong Li
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiaofei Sun
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kaiqiang Sun
- Department of Orthopedic Surgery, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Fanqi Kong
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Jiang
- Department of Anesthesiology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qingjie Kong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
71
|
Liu W, Wang S, Zhang X, Ke Z, Wen X, Zhao J, Zhuang X, Liao L. Enhanced Cardiomyocyte NLRP3 Inflammasome-Mediated Pyroptosis Promotes d-Galactose-Induced Cardiac Aging. J Am Heart Assoc 2024; 13:e032904. [PMID: 38979831 PMCID: PMC11292767 DOI: 10.1161/jaha.123.032904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 06/03/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Cardiac aging represents an independent risk factor for aging-associated cardiovascular diseases. Although evidence suggests an association between NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome formation and numerous cardiovascular diseases, its role in cardiac aging remains largely unclear. METHODS AND RESULTS The longevity of mice with wild-type and NLRP3 knockout (NLRP3-/-) genotypes was assessed, with or without d-galactose treatment. Cardiac function was evaluated using echocardiography, and cardiac histopathology was examined through hematoxylin and eosin and Masson's trichrome staining. Senescence-associated β-galactosidase (SA-β-gal) staining was employed to detect cardiac aging. Western blotting was used to assess aging-related proteins (p53, p21) and pyroptosis-related proteins. Additionally, dihydroethidium staining, lactate dehydrogenase release, and interleukin-1β ELISA assays were performed, along with measurements of total superoxide dismutase and malondialdehyde levels. In vitro, H9c2 cells were exposed to d-galactose for 24 hours in the absence or presence of N-acetyl-l-cysteine (reactive oxygen species inhibitor), BAY-117082 (nuclear factor κ-light-chain enhancer of activated B cells inhibitor), MCC950 (NLRP3 inhibitor), and VX-765 (Caspase-1 inhibitor). Immunofluorescence staining was employed to detect p53, gasdermin D, and apoptosis-associated speck-like protein proteins. Intracellular reactive oxygen species levels were assessed using fluorescence microscopy and flow cytometry. Senescence-associated β-galactosidase staining and Western blotting were also employed in vitro for the same purpose. The results showed that NLRP3 upregulation was implicated in aging and cardiovascular diseases. Inhibition of NLRP3 extended life span, mitigated the aging phenotype, improved cardiac function and blood pressure, ameliorated lipid metabolism abnormalities, inhibited pyroptosis in cardiomyocytes, and ultimately alleviated cardiac aging. In vitro, the inhibition of reactive oxygen species, nuclear factor κ-light-chain enhancer of activated B cells, NLRP3, or caspase-1 attenuated NLRP3 inflammasome-mediated pyroptosis. CONCLUSIONS The reactive oxygen species/nuclear factor κ-light-chain enhancer of activated B cells/NLRP3 signaling pathway loop contributes to d-galactose-treated cardiomyocyte senescence and cardiac aging.
Collapse
Affiliation(s)
- Wen‐bin Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Sui‐sui Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
- Department of Nuclear MedicineThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangdongChina
| | - Xu Zhang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Ze‐zhi Ke
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Xiu‐yun Wen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Jie Zhao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| | - Xiao‐dong Zhuang
- Cardiology DepartmentThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangdongPeople’s Republic of China
| | - Li‐zhen Liao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive SubstancesGuangdong Pharmaceutical University, Guangzhou Higher Education Mega CenterGuangzhouGuangdongChina
- School of Health ScienceGuangdong Pharmaceutical UniversityGuangzhouPeople’s Republic of China
| |
Collapse
|
72
|
Ni L, Yang L, Lin Y. Recent progress of endoplasmic reticulum stress in the mechanism of atherosclerosis. Front Cardiovasc Med 2024; 11:1413441. [PMID: 39070554 PMCID: PMC11282489 DOI: 10.3389/fcvm.2024.1413441] [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: 05/01/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
The research progress of endoplasmic reticulum (ER) stress in atherosclerosis (AS) is of great concern. The ER, a critical cellular organelle, plays a role in important biological processes including protein synthesis, folding, and modification. Various pathological factors may cause ER stress, and sustained or excessive ER stress triggers the unfolded protein response, ultimately resulting in apoptosis and disease. Recently, researchers have discovered the importance of ER stress in the onset and advancement of AS. ER stress contributes to the occurrence of AS through different pathways such as apoptosis, inflammatory response, oxidative stress, and autophagy. Therefore, this review focuses on the mechanisms of ER stress in the development of AS and related therapeutic targets, which will contribute to a deeper understanding of the disease's pathogenesis and provide novel strategies for preventing and treating AS.
Collapse
Affiliation(s)
| | | | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
73
|
Wang J, Li Y, Li L, Liang H, Ye H, Kang P, Li Z, Yu Y, Gao Q. Effect of NLRP3 gene knockdown on pyroptosis and ferroptosis in diabetic cardiomyopathy injury. BMC Cardiovasc Disord 2024; 24:351. [PMID: 38987672 PMCID: PMC11234732 DOI: 10.1186/s12872-024-04010-x] [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: 12/30/2023] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a chronic disease caused by diabetes mellitus, which is recognized as a worldwide challenging disease. This study aimed to investigate the role and the potential mechanism of knocking down the NACHT-, LRR- and PYD domains-containing protein 3 (NLRP3), an inflammasome associated with onset and progression of various diseases, on high glucose or diabetes -induced cardiac cells pyroptosis and ferroptosis, two regulated non-necrosis cell death modalities discovered recent years. In the present study, both in vivo and in vitro studies were conducted simultaneously. Diabetic rats were induced by 55 mg/kg intraperitoneal injection of streptozotocin (STZ). Following the intraperitoneal injection of MCC950 (10 mg/kg), On the other hand, the DCM model in H9C2 cardiac cells was simulated with 35 mmol/L glucose and a short hairpin RNA vector of NLRP3 were transfected to cells. The results showed that in vivo study, myocardial fibers were loosely arranged and showed inflammatory cell infiltration, mitochondrial cristae were broken and the GSDMD-NT expression was found notably increased in the DM group, while the protein expressions of xCT and GPX4 was significantly decreased, both of which were reversed by MCC950. High glucose reduced the cell viability and ATP level in vitro, accompanied by an increase in LDH release. All of the above indicators were reversed after NLRP3 knockdown compared with the HG treated alone. Moreover, the protein expressions of pyroptosis- and ferroptosis-related fators were significantly decreased or increased, consistent with the results shown by immunofluorescence. Furthermore, the protective effects of NLRP3 knockdown against HG were reversed following the mtROS agonist rotenone (ROT) treatment. In conclusion, inhibition of NLRP3 suppressed DM-induced myocardial injury. Promotion of mitochondrial ROS abolished the protective effect of knockdown NLRP3, and induced the happening of pyroptosis and ferroptosis. These findings may present a novel therapeutic underlying mechanism for clinical diabetes-induced myocardial injury treatment.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Anatomy, Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
| | - Yuping Li
- Department of Physiology, Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
| | - Lu Li
- Department of Physiology, Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
| | - Huan Liang
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Department of Graduate Studies, Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, P.R. China
| | - Hongwei Ye
- Department of Physiology, Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
| | - Pinfang Kang
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, P.R. China
| | - Zhenghong Li
- Department of Physiology, Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
| | - Ying Yu
- Department of Physiology, Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China
| | - Qin Gao
- Department of Physiology, Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China.
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, 233000, Anhui, P.R. China.
| |
Collapse
|
74
|
Ge M, Jin L, Cui C, Han Y, Li H, Gao X, Li G, Yu H, Zhang B. Dl-3-n-butylphthalide improves stroke outcomes after focal ischemic stroke in mouse model by inhibiting the pyroptosis-regulated cell death and ameliorating neuroinflammation. Eur J Pharmacol 2024; 974:176593. [PMID: 38636800 DOI: 10.1016/j.ejphar.2024.176593] [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: 01/30/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Recent studies have highlighted the involvement of pyroptosis-mediated cell death and neuroinflammation in ischemic stroke (IS) pathogenesis. DL-3-n-butylphthalide (NBP), a synthesized compound based on an extract from seeds of Apium graveolens, possesses a broad range of biological effects. However, the efficacy and the underlying mechanisms of NBP in IS remain contentious. Herein, we investigated the therapeutic effects of NBP and elucidated its potential mechanisms in neuronal cell pyroptosis and microglia inflammatory responses. Adult male mice underwent permanent distal middle cerebral artery occlusion (dMCAO), followed by daily oral gavage of NBP (80 mg/kg) for 1, 7, or 21 consecutive days. Gene Expression Omnibus (GEO) dataset of IS patients peripheral blood RNA sequencing was analyzed to identify differentially expressed pyroptosis-related genes (PRGs) during the ischemic process. Our results suggested that NBP treatment effectively alleviated brain ischemic damage, resulting in decreased neurological deficit scores, reduced infarct volume, and improved neurological and behavioral functions. RNA sequence data from human unveiled upregulated PRGs in IS. Subsequently, we observed that NBP downregulated pyroptosis-associated markers at days 7 and 21 post-modeling, at both the protein and mRNA levels. Additionally, NBP suppressed the co-localization of pyroptosis markers with neuronal cells to variable degrees and simultaneously mitigated the accumulation of activated microglia. Overall, our data provide novel evidence that NBP treatment significantly attenuates ischemic brain damage and promotes recovery of neurological function in the early and recovery phases after IS, probably by negatively regulating the pyroptosis cell death of neuronal cells and inhibiting toxic neuroinflammation in the central nervous system.
Collapse
Affiliation(s)
- Mengru Ge
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Lingting Jin
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Can Cui
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yingying Han
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hongxia Li
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xue Gao
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Gang Li
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hongxiang Yu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Bei Zhang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
75
|
Norouzi A, Dehghani T, Eftekhar E. Water-pipe Tobacco Components and their Association with Oxidative Stress. ADDICTION & HEALTH 2024; 16:205-212. [PMID: 39439856 PMCID: PMC11491865 DOI: 10.34172/ahj.1487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 05/01/2024] [Indexed: 10/25/2024]
Abstract
Oxidative stress (OS) results from an imbalance between the formation and detoxification of reactive species. Although reactive species at low or moderate levels play numerous physiological roles, high concentrations can lead to disturbances in signaling and metabolic pathways and cause different metabolic, chronic, and age-related disorders. Several endogenous and exogenous processes may lead to the formation of reactive species. The severity of OS can be reduced with the help of antioxidants. Tobacco is one of the most important environmental factors contributing to reactive species production. After cigarette smoking, water-pipe tobacco (WPT) smoking is ranked as the second most popular tobacco product. Its popularity is proliferating due to flavored products, social acceptability, etc. However, studies have shown that WPT smoking is associated with an increased risk of arterial stiffness, ischemic heart disease, and several cancer types. In this study, we aimed to review the most recent evidence on WPT smoking constituents and their association with OS.
Collapse
Affiliation(s)
- Aida Norouzi
- Tobacco and Health Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Tahereh Dehghani
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ebrahim Eftekhar
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
76
|
Zhao P, Zhu J, Bai L, Ma W, Li F, Zhang C, Zhao L, Wang L, Zhang S. Neutrophil extracellular traps induce pyroptosis of pulmonary microvascular endothelial cells by activating the NLRP3 inflammasome. Clin Exp Immunol 2024; 217:89-98. [PMID: 38517050 PMCID: PMC11188539 DOI: 10.1093/cei/uxae028] [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: 11/18/2023] [Revised: 02/27/2024] [Accepted: 03/20/2024] [Indexed: 03/23/2024] Open
Abstract
Excessive formation of neutrophil extracellular traps (NETs) may lead to myositis-related interstitial lung disease (ILD). There is evidence that NETs can directly injure vascular endothelial cells and play a pathogenic role in the inflammatory exudation of ILD. However, the specific mechanism is unclear. This study aimed to investigate the specific mechanism underlying NET-induced injury to human pulmonary microvascular endothelial cells (HPMECs). HPMECs were stimulated with NETs (200 ng/ml) in vitro. Cell death was detected by propidium iodide staining. The morphological changes of the cells were observed by transmission electron microscopy (TEM). Pyroptosis markers were detected by western blot, immunofluorescence, and quantitative real-time polymerase chain reaction, and the related inflammatory factor Interleukin-1β (IL-1β) was verified by enzyme-linked immunosorbent assay (ELISA). Compared with the control group, HPMECs mortality increased after NET stimulation, and the number of pyroptosis vacuoles in HPMECs was further observed by TEM. The pulmonary microvascular endothelial cells (PMECs) of the experimental autoimmune myositis mouse model also showed a trend of pyroptosis in vivo. Cell experiment further confirmed the significantly high expression of the NLRP3 inflammasome and pyroptosis-related markers, including GSDMD and inflammatory factor IL-1β. Pretreated with the NLRP3 inhibitor MCC950, the activation of NLRP3 inflammasome and pyroptosis of HPMECs were effectively inhibited. Our study confirmed that NETs promote pulmonary microvascular endothelial pyroptosis by activating the NLRP3 inflammasome, suggesting that NETs-induced pyroptosis of PMECs may be a potential pathogenic mechanism of inflammatory exudation in ILD.
Collapse
Affiliation(s)
- Peipei Zhao
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jiarui Zhu
- Cui Ying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Ling Bai
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Wenlan Ma
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Feifei Li
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Cen Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Liangtao Zhao
- Cui Ying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Liuyang Wang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
77
|
Ji F, Shi C, Shu Z, Li Z. Nanomaterials Enhance Pyroptosis-Based Tumor Immunotherapy. Int J Nanomedicine 2024; 19:5545-5579. [PMID: 38882539 PMCID: PMC11178094 DOI: 10.2147/ijn.s457309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Abstract
Pyroptosis, a pro-inflammatory and lytic programmed cell death pathway, possesses great potential for antitumor immunotherapy. By releasing cellular contents and a large number of pro-inflammatory factors, tumor cell pyroptosis can promote dendritic cell maturation, increase the intratumoral infiltration of cytotoxic T cells and natural killer cells, and reduce the number of immunosuppressive cells within the tumor. However, the efficient induction of pyroptosis and prevention of damage to normal tissues or cells is an urgent concern to be addressed. Recently, a wide variety of nanoplatforms have been designed to precisely trigger pyroptosis and activate the antitumor immune responses. This review provides an update on the progress in nanotechnology for enhancing pyroptosis-based tumor immunotherapy. Nanomaterials have shown great advantages in triggering pyroptosis by delivering pyroptosis initiators to tumors, increasing oxidative stress in tumor cells, and inducing intracellular osmotic pressure changes or ion imbalances. In addition, the challenges and future perspectives in this field are proposed to advance the clinical translation of pyroptosis-inducing nanomedicines.
Collapse
Affiliation(s)
- Fujian Ji
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Chunyu Shi
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhenbo Shu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhongmin Li
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
78
|
Li Y, Zhang W, Cai Y, Yang D. Ginsenoside Rb2 Inhibits the Pyroptosis in Myocardial Ischemia Progression Through Regulating the SIRT1 Mediated Deacetylation of ASC. Biochem Genet 2024:10.1007/s10528-024-10846-x. [PMID: 38831231 DOI: 10.1007/s10528-024-10846-x] [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: 10/19/2023] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Myocardial ischemic (MI) injury is a common cardiovascular disease, and the potential therapeutic effects of ginsenoside Rb2 (Rb2) have been lately the focus of interest. Therefore, this study aimed to investigate the effects of Rb2 on pyroptosis of cardiomyocytes in MI progression. An in vitro MI model was developed by subjecting rat's cardiomyocytes (H9c2) to hypoxia/reoxygenation (H/R). The cell viability was determined by CCK-8 assay, while cell death was analyzed by propidium iodide staining. Similarly, pyroptosis-related protein levels and acetylation levels of apoptosis-associated speck-like protein containing a CARD (ASC) were detected by western blotting, and the relationship between Sirtuin 1 (SIRT1) and ASC was confirmed by co-immunoprecipitation (Co-IP) assay. Moreover, hematoxylin-eosin (H&E) and triphenyl tetrazolium chloride staining were used to study pathological structure and infarct size. It was found that post-Rb2 treatment significantly increased the cell viability and decreased the cell death and lactic dehydrogenase release, while the increased gasdermin D-N, NOD-like receptor thermal protein domain-associated protein 3, ASC, and cleaved-caspase-1 protein levels were significantly decreased in H/R-stimulated H9c2 cells. Moreover, the acetylation levels of H92c cells were decreased post-Rb2 treatment via increasing SIRT1 levels, while knocking down SIRT1, translated into an increase in ASC acetylation levels, leading to the increase in ASC protein stability and expressions. Additionally, the Rb2 effects on pyroptosis in H/R-stimulated H92c cells were reversed by overexpressing ASC, while reduced myocardial tissue damage was observed in MI rats following in vivo Rb2 treatment. Rb2 treatment inhibited pyroptosis in MI progression by decreasing the ASC levels. Mechanistically, Rb2 treatment increased the SIRT1 levels, further increasing the acetylation levels of ASC and decreasing the protein stability of ASC.
Collapse
Affiliation(s)
- Yuning Li
- Department of Pharmacy, The 921, Hospital of Joint Logistic Support Force of PLA, No.1 Hongshan Bridge, Changsha, 410003, China.
| | - Wenhua Zhang
- Department of Pediatrics, The 3, Hospital of Changsha, Changsha, China
| | - Yamin Cai
- Department of Pharmacy, The 921, Hospital of Joint Logistic Support Force of PLA, No.1 Hongshan Bridge, Changsha, 410003, China
| | - Dong Yang
- Clinical Laboratory, The 921, Hospital of Joint Logistic Support Force of PLA, Changsha, China
| |
Collapse
|
79
|
Liu M, Zeng C, Zhang Y, Xin Y, Deng S, Hu X. Protective role of hydrogen sulfide against diabetic cardiomyopathy by inhibiting pyroptosis and myocardial fibrosis. Biomed Pharmacother 2024; 175:116613. [PMID: 38657502 DOI: 10.1016/j.biopha.2024.116613] [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: 02/12/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) contributes significantly to the heightened mortality rate observed among diabetic patients, with myocardial fibrosis (MF) being a pivotal element in the disease's progression. Hydrogen sulfide (H2S) has been shown to mitigate MF, but the specific underlying mechanisms have yet to be thoroughly understood. A connection has been established between the evolution of DCM and the incidence of cardiomyocyte pyroptosis. Our research offers insights into H2S protective impact and its probable mode of action against DCM, analyzed through the lens of MF. In this study, a diabetic rat model was developed using intraperitoneal injections of streptozotocin (STZ), and hyperglycemia-stimulated cardiomyocytes were employed to replicate the cellular environment of DCM. There was a marked decline in the expression of cystathionine γ-lyase (CSE), a catalyst for H2S synthesis, in both the STZ-induced diabetic rats and hyperglycemia-stimulated cardiomyocytes. Experimental results in vivo indicated that H2S ameliorates MF and enhances cardiac functionality in diabetic rats by mitigating cardiomyocyte pyroptosis. In vitro assessments highlighted the induction of cardiomyocyte pyroptosis and the subsequent decline in cell viability under hyperglycemic conditions. However, the administration of sodium hydrosulfide (NaHS) curtailed cardiomyocyte pyroptosis and augmented cell viability. In contrast, propargylglycine (PAG), a CSE inhibitor, reversed the effects rendered by NaHS administration. Additional exploration indicated that the mitigating effect of H2S on cardiomyocyte pyroptosis is modulated through the ROS/NLRP3 pathway. In essence, our findings corroborate the potential of H2S in alleviating MF in diabetic subjects. This therapeutic effect is likely attributable to the regulation of cardiomyocyte pyroptosis via the ROS/NLRP3 pathway. This discovery furnishes a prospective therapeutic target for the amelioration and management of MF associated with diabetes.
Collapse
Affiliation(s)
- Maojun Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Cheng Zeng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Ying Xin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Simin Deng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Xinqun Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China.
| |
Collapse
|
80
|
Lv Y, Jiang Z, Zhou W, Yang H, Jin G, Wang D, Kong C, Qian Z, Gu Y, Chen S, Zhu L. Low-Shear Stress Promotes Atherosclerosis via Inducing Endothelial Cell Pyroptosis Mediated by IKKε/STAT1/NLRP3 Pathway. Inflammation 2024; 47:1053-1066. [PMID: 38315275 PMCID: PMC11147929 DOI: 10.1007/s10753-023-01960-w] [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: 11/04/2023] [Revised: 12/11/2023] [Accepted: 12/25/2023] [Indexed: 02/07/2024]
Abstract
Atherosclerosis is initiated by vascular endothelial dysfunction, and low-shear stress (LSS) of blood flow is a key factor leading to endothelial dysfunction. Growing evidence suggests that endothelial cell pyroptosis plays an important role in the development of atherosclerosis. Studies have shown that low-shear stress can induce endothelial cell pyroptosis, but the exact mechanism remains unclear. Our experiments demonstrated that low-shear stress induced endothelial cell pyroptosis and the phosphorylation of IκB kinase ε (IKKε). IKKε knockdown not only significantly attenuated atherosclerosis lesions of aortic arch areas in ApoE-/- mice fed with high cholesterol diets, but also markedly reduced endothelial cell pyroptosis and NLRP3 expression triggered by low-shear stress. Further mechanism studies showed that IKKε promoted the expression of NLRP3 via activating signal transducer and activator of transcription 1 (STAT1) and the subsequent binding of STAT1 to NLRP3 promoter region. These results suggest that low-shear stress plays a pro-atherosclerotic role by promoting endothelial cell pyroptosis through the IKKε/STAT1/NLRP3 pathway, which provides new insights into the formation of atherosclerosis.
Collapse
MESH Headings
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Pyroptosis/immunology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- I-kappa B Kinase/genetics
- I-kappa B Kinase/metabolism
- Phosphorylation/immunology
- Mice, Knockout, ApoE
- Gene Knockdown Techniques
- Human Umbilical Vein Endothelial Cells
- Cholesterol, Dietary/administration & dosage
- Cholesterol, Dietary/adverse effects
- Humans
- Animals
- Mice
- Mice, Inbred C57BL
- Signal Transduction/immunology
- STAT1 Transcription Factor/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Stress, Mechanical
- Aorta/cytology
- Aorta/immunology
- Aorta/metabolism
- Aorta/pathology
Collapse
Affiliation(s)
- Yifei Lv
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Zihao Jiang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Wenying Zhou
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Hongfeng Yang
- Department of Intensive Care Unit, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Guozhen Jin
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Dongchen Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Chaohua Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Zhiyuan Qian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China.
| | - Linlin Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210000, China.
| |
Collapse
|
81
|
Huang J, Dong S, Wu Y, Yi H, Zhang W, Ai X. Sirtuin 6 Deacetylates Apoptosis-Associated Speck-Like Protein (ASC) to Inhibit Endothelial Cell Pyroptosis in Atherosclerosis. Int Heart J 2024; 65:466-474. [PMID: 38749754 DOI: 10.1536/ihj.23-334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Endothelial cell dysfunction is the main pathology of atherosclerosis (AS). Sirtuin 6 (SIRT6), a deacetylase, is involved in AS progression. This study aimed to investigate the impacts of SIRT6 on the pyroptosis of endothelial cells and its underlying mechanisms. ApoE-/- mice were fed a high-fat diet (HFD) to establish the AS mouse model, atherosclerotic lesions were evaluated using oil red O staining, and blood lipids and inflammatory factors were measured using corresponding kits. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to establish the cell model, and pyroptosis was evaluated by flow cytometry, ELISA, and western blot. Immunoprecipitation (IP), co-IP, western blot, and immunofluorescence were used to detect the molecular mechanisms. The results showed that SIRT6 expression was downregulated in the blood of HFD-induced mice and ox-LDL-induced HUVECs. Overexpression of SIRT6 reduced atherosclerotic lesions, blood lipids, and inflammation in vivo and suppressed pyroptosis of HUVECs in vitro. Moreover, SIRT6 interacted with ASC to inhibit the acetylation of ASC, thus, reducing the interaction between ASC and NLRP3. Moreover, SIRT6 inhibits endothelial cell pyroptosis in the aortic roots of mice by deacetylating ASC. In conclusion, SIRT6 deacetylated ASC to inhibit its interaction with NLRP3 and then suppressed pyroptosis of endothelial cells, thus, decelerating the progression of AS. The findings provide new insights into the function of SIRT6 in AS.
Collapse
Affiliation(s)
- Jian Huang
- Department of Vascular and Interventional Radiology, The Affiliated Suzhou Hospital of Nanjing Medical University
| | - Shuilin Dong
- Hepatic Surgery Center, Vascular Surgery, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital
| | - Yanhui Wu
- Hepatic Surgery Center, Vascular Surgery, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital
| | - Huiming Yi
- Department of Medical Ultrasound, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital
| | - Wei Zhang
- Department of Medical Ultrasound, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital
| | - Xi Ai
- Department of General Surgery, Huazhong University of Science and Technology, Tongji Medical College, Tongji Hospital
| |
Collapse
|
82
|
Sun Z, Lv R, Zhao Y, Cai Z, Si X, Zhang Q, Liu X. Communications between Neutrophil-Endothelial Interaction in Immune Defense against Bacterial Infection. BIOLOGY 2024; 13:374. [PMID: 38927254 PMCID: PMC11200680 DOI: 10.3390/biology13060374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
The endothelial barrier plays a critical role in immune defense against bacterial infection. Efficient interactions between neutrophils and endothelial cells facilitate the activation of both cell types. However, neutrophil activation can have dual effects, promoting bacterial clearance on one hand while triggering inflammation on the other. In this review, we provide a detailed overview of the cellular defense progression when neutrophils encounter bacteria, focusing specifically on neutrophil-endothelial interactions and endothelial activation or dysfunction. By elucidating the underlying mechanisms of inflammatory pathways, potential therapeutic targets for inflammation caused by endothelial dysfunction may be identified. Overall, our comprehensive understanding of neutrophil-endothelial interactions in modulating innate immunity provides deeper insights into therapeutic strategies for infectious diseases and further promotes the development of antibacterial and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Zhigang Sun
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China; (Z.S.); (Y.Z.); (Z.C.); (X.S.)
| | - Ruoyi Lv
- Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China;
- Beijing Traditional Chinese Veterinary Engineering Center, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China
| | - Yanxin Zhao
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China; (Z.S.); (Y.Z.); (Z.C.); (X.S.)
- Beijing Traditional Chinese Veterinary Engineering Center, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China
| | - Ziwen Cai
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China; (Z.S.); (Y.Z.); (Z.C.); (X.S.)
| | - Xiaohui Si
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China; (Z.S.); (Y.Z.); (Z.C.); (X.S.)
| | - Qian Zhang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China; (Z.S.); (Y.Z.); (Z.C.); (X.S.)
- Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China;
| | - Xiaoye Liu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China; (Z.S.); (Y.Z.); (Z.C.); (X.S.)
- Animal Science and Technology College, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China;
- Beijing Traditional Chinese Veterinary Engineering Center, Beijing University of Agriculture, No. 7 Beinong Road, Changping, Beijing 102206, China
| |
Collapse
|
83
|
Li S, Gu X, Zhang M, Jiang Q, Xu T. Di (2-ethylhexyl) phthalate and polystyrene microplastics co-exposure caused oxidative stress to activate NF-κB/NLRP3 pathway aggravated pyroptosis and inflammation in mouse kidney. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 926:171817. [PMID: 38513858 DOI: 10.1016/j.scitotenv.2024.171817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Polystyrene microplastic (PS-MPs) contamination has become a worldwide hotspot of concern, and its entry into organisms can cause oxidative stress resulting in multi-organ damage. The plasticizer di (2-ethylhexyl) phthalate (DEHP) is a common endocrine disruptor, these two environmental toxins often occur together, but their combined toxicity to the kidney and its mechanism of toxicity are unknown. Therefore, in this study, we established PS-MPS and/or DEHP-exposed mouse models. The results showed that alone exposure to both PS-MPs and DEHP caused inflammatory cell infiltration, cell membrane rupture, and content spillage in kidney tissues. There were also down-regulation of antioxidant enzyme levels, increased ROS content, activated of the NF-κB pathway, stimulated the levels of heat shock proteins (HSPs), pyroptosis, and inflammatory associated factors. Notably, the co-exposure group showed greater toxicity to kidney tissues, the cellular assay further validated these results. The introduction of the antioxidant n-acetylcysteine (NAC) and the NLRP3 inhibitor (MCC950) could mitigate the changes in the above measures. In summary, co-exposure of PS-MPs and DEHP induced oxidative stress that activated the NF-κB/NLRP3 pathway and aggravated kidney pyroptosis and inflammation, as well as that HSPs are also involved in this pathologic injury process. This study not only enriched the nephrotoxicity of plasticizers and microplastics, but also provided new insights into the toxicity mechanisms of multicomponent co-pollution in environmental.
Collapse
Affiliation(s)
- Shanshan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xuedie Gu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Muyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Qihang Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tong Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China..
| |
Collapse
|
84
|
Park HS, Lee BC, Chae DH, Yu A, Park JH, Heo J, Han MH, Cho K, Lee JW, Jung JW, Dunbar CE, Oh MK, Yu KR. Cigarette smoke impairs the hematopoietic supportive property of mesenchymal stem cells via the production of reactive oxygen species and NLRP3 activation. Stem Cell Res Ther 2024; 15:145. [PMID: 38764093 PMCID: PMC11103961 DOI: 10.1186/s13287-024-03731-2] [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: 01/14/2024] [Accepted: 04/10/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) play important roles in tissue homeostasis by providing a supportive microenvironmental niche for the hematopoietic system. Cigarette smoking induces systemic abnormalities, including an impeded recovery process after hematopoietic stem cell transplantation. However, the role of cigarette smoking-mediated alterations in MSC niche function have not been investigated. METHODS In the present study, we investigated whether exposure to cigarette smoking extract (CSE) disrupts the hematopoietic niche function of MSCs, and pathways impacted. To investigate the effects on bone marrow (BM)-derived MSCs and support of hematopoietic stem and progenitor cells (HSPCs), mice were repeatedly infused with the CSE named 3R4F, and hematopoietic stem and progenitor cells (HSPCs) supporting function was determined. The impact of 3R4F on MSCs at cellular level were screened by bulk-RNA sequencing and subsequently validated through qRT-PCR. Specific inhibitors were treated to verify the ROS or NLRP3-specific effects, and the cells were then transplanted into the animal model or subjected to coculture with HSPCs. RESULTS Both direct ex vivo and systemic in vivo MSC exposure to 3R4F resulted in impaired engraftment in a humanized mouse model. Furthermore, transcriptomic profile analysis showed significantly upregulated signaling pathways related to reactive oxygen species (ROS), inflammation, and aging in 3R4F-treated MSCs. Notably, ingenuity pathway analysis revealed the activation of NLRP3 inflammasome signaling pathway in 3R4F-treated MSCs, and pretreatment with the NLRP3 inhibitor MCC950 rescued the HSPC-supporting ability of 3R4F-treated MSCs. CONCLUSION In conclusion, these findings indicate that exposure to CSE reduces HSPCs supportive function of MSCs by inducing robust ROS production and subsequent NLRP3 activation.
Collapse
Affiliation(s)
- Hyun Sung Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Byung-Chul Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Dong-Hoon Chae
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Aaron Yu
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Jae Han Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Jiyoung Heo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Myoung Hee Han
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Keonwoo Cho
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Joong Won Lee
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Korea
| | - Ji-Won Jung
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Korea
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mi-Kyung Oh
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Kyung-Rok Yu
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
85
|
Chang X, Wang B, Zhao Y, Deng B, Liu P, Wang Y. The role of IFI16 in regulating PANoptosis and implication in heart diseases. Cell Death Discov 2024; 10:204. [PMID: 38693141 PMCID: PMC11063201 DOI: 10.1038/s41420-024-01978-5] [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: 12/13/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
Interferon Gamma Inducible Protein 16 (IFI16) belongs to the HIN-200 protein family and is pivotal in immunological responses. Serving as a DNA sensor, IFI16 identifies viral and aberrant DNA, triggering immune and inflammatory responses. It is implicated in diverse cellular death mechanisms, such as pyroptosis, apoptosis, and necroptosis. Notably, these processes are integral to the emergent concept of PANoptosis, which encompasses cellular demise and inflammatory pathways. Current research implies a significant regulatory role for IFI16 in PANoptosis, particularly regarding cardiac pathologies. This review delves into the complex interplay between IFI16 and PANoptosis in heart diseases, including atherosclerosis, myocardial infarction, heart failure, and diabetic cardiomyopathy. It synthesizes evidence of IFI16's impact on PANoptosis, with the intention of providing novel insights for therapeutic strategies targeting heart diseases.
Collapse
Affiliation(s)
- Xindi Chang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bei Wang
- Department of Emergency, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Yingli Zhao
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bing Deng
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| | - Yiru Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| |
Collapse
|
86
|
Chandy M, Hill T, Jimenez-Tellez N, Wu JC, Sarles SE, Hensel E, Wang Q, Rahman I, Conklin DJ. Addressing Cardiovascular Toxicity Risk of Electronic Nicotine Delivery Systems in the Twenty-First Century: "What Are the Tools Needed for the Job?" and "Do We Have Them?". Cardiovasc Toxicol 2024; 24:435-471. [PMID: 38555547 PMCID: PMC11485265 DOI: 10.1007/s12012-024-09850-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Cigarette smoking is positively and robustly associated with cardiovascular disease (CVD), including hypertension, atherosclerosis, cardiac arrhythmias, stroke, thromboembolism, myocardial infarctions, and heart failure. However, after more than a decade of ENDS presence in the U.S. marketplace, uncertainty persists regarding the long-term health consequences of ENDS use for CVD. New approach methods (NAMs) in the field of toxicology are being developed to enhance rapid prediction of human health hazards. Recent technical advances can now consider impact of biological factors such as sex and race/ethnicity, permitting application of NAMs findings to health equity and environmental justice issues. This has been the case for hazard assessments of drugs and environmental chemicals in areas such as cardiovascular, respiratory, and developmental toxicity. Despite these advances, a shortage of widely accepted methodologies to predict the impact of ENDS use on human health slows the application of regulatory oversight and the protection of public health. Minimizing the time between the emergence of risk (e.g., ENDS use) and the administration of well-founded regulatory policy requires thoughtful consideration of the currently available sources of data, their applicability to the prediction of health outcomes, and whether these available data streams are enough to support an actionable decision. This challenge forms the basis of this white paper on how best to reveal potential toxicities of ENDS use in the human cardiovascular system-a primary target of conventional tobacco smoking. We identify current approaches used to evaluate the impacts of tobacco on cardiovascular health, in particular emerging techniques that replace, reduce, and refine slower and more costly animal models with NAMs platforms that can be applied to tobacco regulatory science. The limitations of these emerging platforms are addressed, and systems biology approaches to close the knowledge gap between traditional models and NAMs are proposed. It is hoped that these suggestions and their adoption within the greater scientific community will result in fresh data streams that will support and enhance the scientific evaluation and subsequent decision-making of tobacco regulatory agencies worldwide.
Collapse
Affiliation(s)
- Mark Chandy
- Robarts Research Institute, Western University, London, N6A 5K8, Canada
| | - Thomas Hill
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Nerea Jimenez-Tellez
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - S Emma Sarles
- Biomedical and Chemical Engineering PhD Program, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Edward Hensel
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Daniel J Conklin
- Division of Environmental Medicine, Department of Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, 580 S. Preston St., Delia Baxter, Rm. 404E, Louisville, KY, 40202, USA.
| |
Collapse
|
87
|
Hu Q, Zhang R, Dong X, Yang D, Yu W, Du Q. Huperzine A ameliorates neurological deficits after spontaneous subarachnoid hemorrhage through endothelial cell pyroptosis inhibition. Acta Biochim Biophys Sin (Shanghai) 2024; 56:645-656. [PMID: 38529553 DOI: 10.3724/abbs.2024037] [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] [Indexed: 03/27/2024] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH) is a kind of hemorrhagic stroke which causes neurological deficits in survivors. Huperzine A has a neuroprotective effect, but its role in SAH is unclear. Therefore, we explore the effect of Huperzine A on neurological deficits induced by SAH and the related mechanism. In this study, Evans blue assay, TUNEL staining, immunofluorescence, western blot analysis, and ELISA are conducted. We find that Huperzine A can improve neurological deficits and inhibit the apoptosis of nerve cells in SAH rats. Huperzine A treatment can improve the upregulation of brain water content, damage of blood-brain barrier, fibrinogen and matrix metalloprotein 9 expressions and the downregulation of ZO-1 and occludin expressions induced by SAH. Huperzine A inhibit the expressions of proteins involved in pyroptosis in endothelial cells in SAH rats. The increase in MDA content and decrease in SOD activity in SAH rats can be partly reversed by Huperzine A. The ROS inducer H 2O 2 can induce pyroptosis and inhibit the expressions of ZO-1 and occludin in endothelial cells, which can be blocked by Huperzine A. In addition, the increase in the entry of p65 into the nucleus in endothelial cells can be partly reversed by Huperzine A. Huperzine A may delay the damage of blood-brain barrier in SAH rats by inhibiting oxidative stress-mediated pyroptosis and tight junction protein expression downregulation through the NF-κB pathway. Overall, Huperzine A may have clinical value for treating SAH.
Collapse
Affiliation(s)
- Qiang Hu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Rong Zhang
- Medical Examination Center, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
| | - Xiaoqiao Dong
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Dingbo Yang
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Wenhua Yu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310000, China
- Department of Neurosurgery, Nanjing Medical University Affiliated Hangzhou Hospital, Hangzhou First People's Hospital, Hangzhou 310000, China
| |
Collapse
|
88
|
Jiang XS, Liu T, Xia YF, Gan H, Ren W, Du XG. Activation of the Nrf2/ARE signaling pathway ameliorates hyperlipidemia-induced renal tubular epithelial cell injury by inhibiting mtROS-mediated NLRP3 inflammasome activation. Front Immunol 2024; 15:1342350. [PMID: 38720901 PMCID: PMC11076710 DOI: 10.3389/fimmu.2024.1342350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/02/2024] [Indexed: 05/12/2024] Open
Abstract
Dyslipidemia is the most prevalent independent risk factor for patients with chronic kidney disease (CKD). Lipid-induced NLRP3 inflammasome activation in kidney-resident cells exacerbates renal injury by causing sterile inflammation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that modulates the cellular redox balance; however, the exact role of Nrf2 signaling and its regulation of the NLRP3 inflammasome in hyperlipidemia-induced kidney injury are poorly understood. In this study, we demonstrated that activation of the mtROS-NLRP3 inflammasome pathway is a critical contributor to renal tubular epithelial cell (RTEC) apoptosis under hyperlipidemia. In addition, the Nrf2/ARE signaling pathway is activated in renal tubular epithelial cells under hyperlipidemia conditions both in vivo and in vitro, and Nrf2 silencing accelerated palmitic acid (PA)-induced mtROS production, mitochondrial injury, and NLRP3 inflammasome activation. However, the activation of Nrf2 with tBHQ ameliorated mtROS production, mitochondrial injury, NLRP3 inflammasome activation, and cell apoptosis in PA-induced HK-2 cells and in the kidneys of HFD-induced obese rats. Furthermore, mechanistic studies showed that the potential mechanism of Nrf2-induced NLRP3 inflammasome inhibition involved reducing mtROS generation. Taken together, our results demonstrate that the Nrf2/ARE signaling pathway attenuates hyperlipidemia-induced renal injury through its antioxidative and anti-inflammatory effects through the downregulation of mtROS-mediated NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xu-shun Jiang
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Liu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun-feng Xia
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Ren
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-gang Du
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
89
|
Shao X, Zeng W, Wang Q, Liu S, Guo Q, Luo D, Luo Q, Wang D, Wang L, Zhang Y, Diao H, Piao S, Yan M, Guo J. Fufang Zhenzhu Tiaozhi (FTZ) suppression of macrophage pyroptosis: Key to stabilizing rupture-prone plaques. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117705. [PMID: 38219878 DOI: 10.1016/j.jep.2024.117705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Research on the Chinese herbal formula Fufang Zhenzhu Tiaozhi (FTZ) has demonstrated its effectiveness in treating hyperlipidemia and glycolipid metabolic disorders. Additionally, FTZ has shown inhibitory effects on oxidative stress, regulation of lipid metabolism, and reduction of inflammation in these conditions. However, the precise mechanisms through which FTZ modulates macrophage function in atherosclerosis remain incompletely understood. Therefore, this study aims to investigate whether FTZ can effectively stabilize rupture-prone plaques by suppressing macrophage pyroptosis and impeding the development of M1 macrophage polarization in ApoE-/- mice. METHODS To assess the impact of FTZ on macrophage function and atherosclerosis in ApoE-/- mice, we orally administered FTZ at a dosage of 1.2 g/kg body weight daily for 14 weeks. Levels of interleukin-18 and interleukin-1β were quantified using ELISA kits to gauge FTZ's influence on inflammation. Total cholesterol content was measured with a Cholesterol Assay Kit to evaluate FTZ's effect on lipid metabolism. Aortic tissues were stained with Oil Red O, and immunohistochemistry techniques were applied to assess atherosclerotic lesions and plaque stability. To evaluate the effects of FTZ on macrophage pyroptosis and oxidative damage, immunofluorescence staining was utilized. Additionally, we conducted an analysis of protein and mRNA expression levels of NLRP3 inflammasome-related genes and macrophage polarization-related genes using RT-PCR and western blotting techniques. RESULTS This study illustrates the potential therapeutic effectiveness of FTZ in mitigating the severity of atherosclerosis and improving serum lipid profiles by inhibiting inflammation. The observed enhancements in atherosclerosis severity and inflammation can be attributed to the suppression of NLRP3 inflammasome activity and M1 polarization by FTZ. CONCLUSION The current findings indicate that FTZ provides protection against atherosclerosis, positioning it as a promising candidate for novel therapies targeting atherosclerosis and related cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaoqi Shao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Wenru Zeng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Qing Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Suping Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Qiaoling Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Duosheng Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Qingmao Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Dongwei Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lexun Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Yue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Hongtao Diao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Shenghua Piao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Meiling Yan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
90
|
Chung C, Park SY, Huh JY, Kim NH, Shon C, Oh EY, Park YJ, Lee SJ, Kim HC, Lee SW. Fine particulate matter aggravates smoking induced lung injury via NLRP3/caspase-1 pathway in COPD. J Inflamm (Lond) 2024; 21:13. [PMID: 38654364 DOI: 10.1186/s12950-024-00384-z] [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: 10/22/2022] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Exposure to noxious particles, including cigarette smoke and fine particulate matter (PM2.5), is a risk factor for chronic obstructive pulmonary disease (COPD) and promotes inflammation and cell death in the lungs. We investigated the combined effects of cigarette smoking and PM2.5 exposure in patients with COPD, mice, and human bronchial epithelial cells. METHODS The relationship between PM2.5 exposure and clinical parameters was investigated in patients with COPD based on smoking status. Alveolar destruction, inflammatory cell infiltration, and pro-inflammatory cytokines were monitored in the smoking-exposed emphysema mouse model. To investigate the mechanisms, cell viability and death and pyroptosis-related changes in BEAS-2B cells were assessed following the exposure to cigarette smoke extract (CSE) and PM2.5. RESULTS High levels of ambient PM2.5 were more strongly associated with high Saint George's respiratory questionnaire specific for COPD (SGRQ-C) scores in currently smoking patients with COPD. Combined exposure to cigarette smoke and PM2.5 increased mean linear intercept and TUNEL-positive cells in lung tissue, which was associated with increased inflammatory cell infiltration and inflammatory cytokine release in mice. Exposure to a combination of CSE and PM2.5 reduced cell viability and upregulated NLRP3, caspase-1, IL-1β, and IL-18 transcription in BEAS-2B cells. NLRP3 silencing with siRNA reduced pyroptosis and restored cell viability. CONCLUSIONS PM2.5 aggravates smoking-induced airway inflammation and cell death via pyroptosis. Clinically, PM2.5 deteriorates quality of life and may worsen prognosis in currently smoking patients with COPD.
Collapse
Affiliation(s)
- Chiwook Chung
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Department of Pulmonary and Critical Care Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - Suk Young Park
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
| | - Jin-Young Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Chung- Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Republic of Korea
| | - Na Hyun Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
| | - ChangHo Shon
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Efficacy Evaluation Center, WOOJUNGBIO Inc, Hwaseong, Republic of Korea
| | - Eun Yi Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Jun Park
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seon-Jin Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hwan-Cheol Kim
- Department of Occupational and Environmental Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea.
| |
Collapse
|
91
|
Fang M, Xia F, Wang J, Wang C, Teng B, You S, Li M, Chen X, Hu X. The NLRP3 inhibitor, OLT1177 attenuates brain injury in experimental intracerebral hemorrhage. Int Immunopharmacol 2024; 131:111869. [PMID: 38492343 DOI: 10.1016/j.intimp.2024.111869] [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: 10/29/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND AND PURPOSE It has been reported activation of NLRP3 inflammasome after intracerebral hemorrhage (ICH) ictus exacerbates neuroinflammation and brain injury. We hypothesized that inhibition of NLRP3 by OLT1177 (dapansutrile), a novel NLRP3 inflammasome inhibitor, could reduce brain edema and attenuate brain injury in experimental ICH. METHODS ICH was induced by injection of autologous blood into basal ganglia in mice models. Sixty-three C57Bl/6 male mice were randomly grouped into the sham, vehicle, OLT1177 (Dapansutrile, 200 mg/kg intraperitoneally) and treated for consecutive three days, starting from 1 h after ICH surgery. Behavioral test, brain edema, brain water content, blood-brain barrier integrity and vascular permeability, cell apoptosis, and NLRP3 and its downstream protein levels were measured. RESULTS OLT1177 significantly reduced cerebral edema after ICH and contributed to the attenuation of neurological deficits. OLT1177 could preserve blood-brain barrier integrity and lessen vascular leakage. In addition, OLT1177 preserved microglia morphological shift and significantly inhibited the activation of caspase-1 and release of IL-1β. We also found that OLT1177 can protect against neuronal loss in the affected hemisphere. CONCLUSIONS OLT1177 (dapansutrile) could significantly attenuate the brain edema after ICH and effectively alleviate the neurological deficit. This result suggests that the novel NLRP3 inhibitor, OLT1177, might serve as a promising candidate for the treatment of ICH.
Collapse
Affiliation(s)
- Mei Fang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiayan Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengyang Wang
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Bang Teng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shenglan You
- Animal Imaging Core Facilities, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Manrui Li
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiameng Chen
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Xin Hu
- Animal Imaging Core Facilities, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
92
|
Chen Y, Yuan C, Qin W, Yu B, Wei D, Wu P. TMAO promotes vascular endothelial cell pyroptosis via the LPEAT-mitophagy pathway. Biochem Biophys Res Commun 2024; 703:149667. [PMID: 38382362 DOI: 10.1016/j.bbrc.2024.149667] [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: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Trimethylamine N-oxide (TMAO) is a novel risk factor for atherosclerosis, and its underlying regulatory mechanisms are under intensive investigation. Inflammation-related vascular endothelial damage is the major driver in atherogenic process. Pyroptosis, a type of proinflammatory programmed cell death, has been proved to promote the initiation and progression of atherosclerosis. In our study, we found that TMAO triggered endothelial cells excessive mitophagy, thereby facilitating pyroptosis. This process is mediated by the upexpression of phosphatidylethanolamine acyltransferase (LPEAT). These findings provide insights into TMAO-induced vascular endothelial cell damage and suggest that LPEAT may be a valuable target for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yanmei Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Department of Pathology, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Chuchu Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Wenhua Qin
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bo Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Peng Wu
- Hengyang Maternal and Child Health Hospital, Hengyang, 421001, Hunan Province, China; Hunan YueYang Maternal & Child Medicine Health-Care Hospital, Hunan Province Innovative Training Base for Medical Postgraduates, Yueyang, Hunan, 414000, China.
| |
Collapse
|
93
|
Xu T, Chen T, Fang H, Shen X, Shen X, Tang Z, Zhao J. Human Umbilical Cord Mesenchymal Stem Cells Repair Endothelial Injury and Dysfunction by Regulating NLRP3 to Inhibit Endothelial Cell Pyroptosis in Kawasaki Disease. Inflammation 2024; 47:483-502. [PMID: 37948033 DOI: 10.1007/s10753-023-01921-3] [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: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
Vascular endothelial inflammation and endothelial dysfunction are the main causes of endothelial injury in Kawasaki disease (KD). Human umbilical cord-derived mesenchymal stem cells (Huc-MSCs) have multiple functions in immune regulation. This study examined whether Huc-MSCs inhibited endothelial inflammation and improved endothelial function in KD through constructing cell and in vivo animal KD vasculitis models. The pyroptosis factor NOD-like receptor protein 3 (NLRP3) was involved in the inflammatory process in the acute phase of KD. After tail vein injection of Huc-MSCs, inflammatory cell infiltration and the expression of pyroptosis-related proteins in the LCWE-induced KD mouse vasculitis model were significantly reduced. In vitro, NLRP3-dependent pyroptosis successfully induced human umbilical vein endothelial cell (HUVEC) damage. Huc-MSCs effectively increased the abilities of impaired HUVECs to proliferate, migrate, invade, and form vessel-like tubes, while inhibiting their apoptosis, suggesting that Huc-MSCs can reduce inflammation and improve vascular endothelial function by inhibiting the NLRP3-dependent pyroptosis pathway in KD, providing a possibility and novel target for KD endothelial injury and dysfunction.
Collapse
Affiliation(s)
- Ting Xu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Tao Chen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Hao Fang
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xiwei Shen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
- Research Institute of Comparative Medicine, Nantong University, Nantong Jiangsu Province, 226001, China
| | - Xianjuan Shen
- Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Jianmei Zhao
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China.
| |
Collapse
|
94
|
Zhou G, Liu Y, Wu H, Zhang D, Yang Q, Li Y. Research Progress on Histone Deacetylases Regulating Programmed Cell Death in Atherosclerosis. J Cardiovasc Transl Res 2024; 17:308-321. [PMID: 37821683 DOI: 10.1007/s12265-023-10444-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
Histone deacetylases (HDACs) are epigenetic modifying enzyme that is closely related to chromatin structure and gene transcription, and numerous studies have found that HDACs play an important regulatory role in atherosclerosis disease. Apoptosis, autophagy and programmed necrosis as the three typical programmed cell death modalities that can lead to cell loss and are closely related to the developmental process of atherosclerosis. In recent years, accumulating evidence has shown that the programmed cell death mediated by HDACs is increasingly important in the pathophysiology of atherosclerosis. This paper first gives a brief overview of HDACs, the mechanism of programmed cell death, and their role in atherosclerosis, and then further elaborates on the role and mechanism of HDACs in regulating apoptosis, autophagy, and programmed necrosis in atherosclerosis, respectively, to provide new effective measures and theoretical basis for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Gang Zhou
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Yanfang Liu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Hui Wu
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China.
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China.
- Department of Cardiology, Yichang Central People's Hospital, Yiling Road 183, Yichang, 443003, Hubei, China.
| | - Dong Zhang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Qingzhuo Yang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| | - Yi Li
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, China
- Department of Central Experimental Laboratory, Yichang Central People's Hospital, Yichang, 443003, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443003, China
| |
Collapse
|
95
|
Li S, Li Q, Zhou Q, Li S, Wang S, Yao Q, Ouyang C, Liu C, Li M. Attenuating Atherosclerosis through Inhibition of the NF- κB/NLRP3/IL-1 β Pathway-Mediated Pyroptosis in Vascular Smooth Muscle Cells (VSMCs). Cardiovasc Ther 2024; 2024:1506083. [PMID: 39742016 PMCID: PMC10985643 DOI: 10.1155/2024/1506083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 01/03/2025] Open
Abstract
Objective We investigated the effects of resveratrol (Res) and MCC950 on the pyroptosis of vascular smooth muscle cells (VSMCs) and the potential pathway. Methods and Results Compared with the control (Con) group, the atherosclerosis (AS) group showed calcified nodules, which suggested that the calcification medium induced the calcification of VSMCs. VSMCs showed proliferative activity and significantly attenuated calcification under treatment with 10 μmol/L Res. The calcium salt was detected by alizarin red S staining. Res and MCC950 downregulated the calcification, inflammatory, pyroptosis, and transcription factor-related indicators all decreased by RT-qPCR with Western blot and immunofluorescence. The results showed that Res and MCC950 refrained the calcification of VSMCs and that Res has a better effect than MCC950. Plaques and calcium salt deposits were present in the carotid region in the control group. More calcium salt deposits were evident in the plaques of the Par group by HE staining and alizarin red S staining. The calcification indexes BMP2, Runx2, and related indexes declined by immunofluorescence, which showed parthenolide-inhibited AS. The related protein expressions were consistent with the expression of the cell experiments. Conclusion Our data demonstrated that inflammatory response and pyroptosis exacerbate AS and unravel the link between VSMCs and the progression of AS lesions. Res and MCC950 inhibited the calcification of VSMCs by regulating NF-κB/NLRP3/IL-1β signaling axis. P53 can exacerbate the AS lesions by acting on NLRP3 inflammasome and pyroptosis. Our findings supported the clinical applications of Res and MCC950 in VSMCs individuals to counteract pyroptosis and AS, and P53 inhibitors also can be a potential treatment for AS.
Collapse
MESH Headings
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Pyroptosis/drug effects
- Signal Transduction/drug effects
- NF-kappa B/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/drug therapy
- Interleukin-1beta/metabolism
- Resveratrol/pharmacology
- Cells, Cultured
- Furans/pharmacology
- Sulfonamides/pharmacology
- Disease Models, Animal
- Animals
- Anti-Inflammatory Agents/pharmacology
- Male
- Indenes/pharmacology
- Plaque, Atherosclerotic
- Inflammasomes/metabolism
- Humans
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Carotid Artery Diseases/pathology
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/drug therapy
- Carotid Artery Diseases/genetics
Collapse
Affiliation(s)
- Shihuan Li
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Qingjie Li
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Qiaofeng Zhou
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Suqin Li
- School of Basic Medical Science, Hubei University of Science and Technology, Xianning, China
| | - Siqi Wang
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Qing Yao
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Changhan Ouyang
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
| | - Chao Liu
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Mincai Li
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
- School of Basic Medical Science, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
96
|
Liang W, Wei R, Zhu X, Li J, Lin A, Chen J, Wu W, Jie Q. Downregulation of HMGB1 carried by macrophage-derived extracellular vesicles delays atherosclerotic plaque formation through Caspase-11-dependent macrophage pyroptosis. Mol Med 2024; 30:38. [PMID: 38493291 PMCID: PMC10943908 DOI: 10.1186/s10020-023-00753-z] [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: 04/02/2023] [Accepted: 11/02/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Macrophage-derived extracellular vesicle (macrophage-EV) is highly studied for its regulatory role in atherosclerosis (AS). Our current study tried to elucidate the possible role of macrophage-EV loaded with small interfering RNA against high-mobility group box 1 (siHMGB1) affecting atherosclerotic plaque formation. METHODS In silico analysis was performed to find critical factors in mouse atherosclerotic plaque formation. EVs secreted by RAW 264.7 cells were collected by ultracentrifugation and characterized, followed by the preparation of macrophage-EV-loaded siHMGB1 (macrophage-EV/siHMGB1). ApoE-/- mice were used to construct an AS mouse model by a high-fat diet, followed by injection of macrophage-EV/siHMGB1 to assess the in vivo effect of macrophage-EV/siHMGB1 on AS mice. RAW264.7 cells were subjected to ox-LDL, LPS or macrophage-EV/siHMGB1 for analyzing the in vitro effect of macrophage-EV/siHMGB1 on macrophage pyrophosis and inflammation. RESULTS In silico analysis found that HMGB1 was closely related to the development of AS. Macrophage-EV/siHMGB could inhibit the release of HMGB1 from macrophages to outside cells, and the reduced HMGB1 release could inhibit foam cell formation. Besides, macrophage-EV/siHMGB also inhibited the LPS-induced Caspase-11 activation, thus inhibiting macrophage pyroptosis and preventing atherosclerotic plaque formation. CONCLUSION Our results proved that macrophage-EV/siHMGB could inhibit foam cell formation and suppress macrophage pyroptosis, finally preventing atherosclerotic plaque formation in AS mice.
Collapse
Affiliation(s)
- Weijie Liang
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Ruibin Wei
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Xingxing Zhu
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Jinliang Li
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Aiwen Lin
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Jun Chen
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Wen Wu
- Department of Endocrinology, Guangdong Geriatrics Institute, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, No. 106, Zhongshan Second Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Qiang Jie
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China.
| |
Collapse
|
97
|
Ishida M, Sakai C, Kobayashi Y, Ishida T. Cigarette Smoking and Atherosclerotic Cardiovascular Disease. J Atheroscler Thromb 2024; 31:189-200. [PMID: 38220184 PMCID: PMC10918046 DOI: 10.5551/jat.rv22015] [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: 10/31/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024] Open
Abstract
The detrimental effects of cigarette smoking on cardiovascular health, particularly atherosclerosis and thrombosis, are well established, and more detailed mechanisms continue to emerge. As the fundamental pathophysiology of the adverse effects of smoking, endothelial dysfunction, inflammation, and thrombosis are considered to be particularly important. Cigarette smoke induces endothelial dysfunction, leading to impaired vascular dilation and hemostasis regulation. Factors contributing to endothelial dysfunction include reduced bioavailability of nitric oxide, increased levels of superoxide anion, and endothelin release. Chronic inflammation of the vascular wall is a central pathogenesis of smoking-induced atherosclerosis. Smoking systemically elevates inflammatory markers and induces the expression of adhesion molecules and cytokines in various tissues. Pattern recognition receptors and damage-associated molecular patterns play crucial roles in the mechanism underlying smoking-induced inflammation. Smoking-induced DNA damage and activation of innate immunity, such as the NLRP3 inflammasome, cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, and Toll-like receptor 9, are shown to amplify inflammatory cytokine expression. Cigarette smoke-induced oxidative stress and inflammation influence platelet adhesion, aggregation, and coagulation via adhesion molecule upregulation. Furthermore, it affects the coagulation cascade and fibrinolysis balance, causing thrombus formation. Matrix metalloproteinases contribute to plaque vulnerability and atherothrombotic events. The impact of smoking on inflammatory cells and adhesion molecules further intensifies the risk of atherothrombosis. Collectively, exposure to cigarette smoke exerts profound effects on endothelial function, inflammation, and thrombosis, contributing to the development and progression of atherosclerosis and atherothrombotic cardiovascular diseases. Understanding these intricate mechanisms highlights the urgent need for smoking cessation to protect cardiovascular health. This comprehensive review investigates the multifaceted mechanisms through which smoking contributes to these life-threatening conditions.
Collapse
Affiliation(s)
- Mari Ishida
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chiemi Sakai
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yusuke Kobayashi
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
98
|
Gan T, Xing Q, Li N, Deng Z, Pan C, Liu X, Zheng L. Protective Effect of Vitexin Against IL-17-Induced Vascular Endothelial Inflammation Through Keap1/Nrf2-Dependent Signaling Pathway. Mol Nutr Food Res 2024; 68:e2300331. [PMID: 38299432 DOI: 10.1002/mnfr.202300331] [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: 05/23/2023] [Revised: 08/31/2023] [Indexed: 02/02/2024]
Abstract
SCOPE Vitexin, a C-glycosylated flavonoid, is abundant in food sources and has potential health-beneficial properties. However, the targets for its beneficial effects remain largely unknown. This study aims to establish an in vitro cell model of vascular low-grade inflammation and explore the antiinflammatory mechanism of vitexin. METHODS AND RESULTS Low-dose TNFα and IL-17 are combined to establish a cell model of vascular low-grade inflammation. Cell-based studies show that low-dose TNFα (1 ng mL-1) alone has a slight effect, but its combination with IL-17 can potently induce protein expression of inflammatory cytokines, leading to an inflammatory state. However, the vascular inflammation caused by low-dose TNF plus IL-17 does not lead to oxidative stress, and reactive oxygen species (ROS) does not involved in developing this inflammation. Vitexin can be absorbed by human umbilical vein endothelial (HUVEC) cells to increase the Nrf2 protein level and attenuate inflammation. In addition, the antiinflammatory effect of vitexin is blocked by the knockdown of Nrf2. Further localized surface plasmon resonance, drug affinity responsive target stability, and molecular docking demonstrate that vitexin can directly interact with Keap1 to disrupt Keap1-Nrf2 interaction and thus activate Nrf2. Treatment of mice with a bolus oral gavage of vitexin (100 mg kg-1 body weight) or a high-fat diet supplemented with vitexin (5 mg kg-1 body weight per day) for 12 weeks confirms the rapid increase in blood vitexin levels and subsequent incorporation into blood vessels to activate Nrf2 and ameliorate inflammation in vivo. CONCLUSION The findings provide a reliable cell model of vascular low-grade inflammation and indicate Nrf2 protein as the potential target of vitexin to inhibit vascular inflammation.
Collapse
Affiliation(s)
- Ting Gan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, 330047, China
| | - Qian Xing
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, 330047, China
| | - Nan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, 330047, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, 330047, China
- Institute for Advanced Study, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Changxuan Pan
- Inspection and Quarantine and Epidemic Prevention and Control Center of Daxing District Agriculture and Rural Bureau of Beijing, Beijing, 102600, China
| | - Xiaoru Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, 330047, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, 330047, China
| |
Collapse
|
99
|
Zhang T, Pang C, Xu M, Zhao Q, Hu Z, Jiang X, Guo M. The role of immune system in atherosclerosis: Molecular mechanisms, controversies, and future possibilities. Hum Immunol 2024; 85:110765. [PMID: 38369442 DOI: 10.1016/j.humimm.2024.110765] [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: 07/17/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Numerous cardiovascular disorders have atherosclerosis as their pathological underpinning. Numerous studies have demonstrated that, with the aid of pattern recognition receptors, cytokines, and immunoglobulins, innate immunity, represented by monocytes/macrophages, and adaptive immunity, primarily T/B cells, play a critical role in controlling inflammation and abnormal lipid metabolism in atherosclerosis. Additionally, the finding of numerous complement components in atherosclerotic plaques suggests yet again how heavily the immune system controls atherosclerosis. Therefore, it is essential to have a thorough grasp of how the immune system contributes to atherosclerosis. The specific molecular mechanisms involved in the activation of immune cells and immune molecules in atherosclerosis, the controversy surrounding some immune cells in atherosclerosis, and the limitations of extrapolating from relevant animal models to humans were all carefully reviewed in this review from the three perspectives of innate immunity, adaptive immunity, and complement system. This could provide fresh possibilities for atherosclerosis research and treatment in the future.
Collapse
Affiliation(s)
- Tianle Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenxu Pang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Qianqian Zhao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhijie Hu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
100
|
Hong Y, Zhou X, Li Q, Chen J, Wei Y, Long C, Shen L, Zheng X, Li D, Wang X, Yu C, Wu S, Wei G. X-box binding protein 1 caused an imbalance in pyroptosis and mitophagy in immature rats with di-(2-ethylhexyl) phthalate-induced testis toxicity. Genes Dis 2024; 11:935-951. [PMID: 37692514 PMCID: PMC10491871 DOI: 10.1016/j.gendis.2023.02.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/08/2023] [Indexed: 03/29/2023] Open
Abstract
As a widely used plasticizer, di-(2-ethylhexyl) phthalate (DEHP) is known to induce significant testicular injury. However, the potential mechanism and effects of pubertal exposure to DEHP on testis development remain unclear. In vivo, postnatal day (PND) 21 male rats were gavaged with 0, 250, and 500 mg/kg DEHP for ten days. Damage to the seminiferous epithelium and disturbed spermatogenesis were observed after DEHP exposure. Meanwhile, oxidative stress-induced injury and pyroptosis were activated. Both endoplasmic reticulum (ER) stress and mitophagy were involved in this process. Monoethylhexyl phthalate (MEHP) was used as the biometabolite of DEHP in vitro. The GC-1 and GC-2 cell lines were exposed to 0, 100 μM, 200 μM, and 400 μM MEHP for 24 h. Reactive oxygen species (ROS) generation, oxidative stress damage, ER stress, mitophagy, and pyroptosis were significantly increased after MEHP exposure. The ultrastructure of the ER and mitochondria was destroyed. X-box binding protein 1 (XBP1) was observed to be activated and translocated into the nucleus. ROS generation was inhibited by acetylcysteine. The levels of antioxidative stress, ER stress, mitophagy, and pyroptosis were decreased as well. After the administration of the ER stress inhibitor 4-phenyl-butyric acid, both mitophagy and pyroptosis were inhibited. Toyocamycin-induced XBP1 down-regulation decreased the levels of mitophagy and pyroptosis. The equilibrium between pyroptosis and mitophagy was disturbed by XBP1 accumulation. In summary, our findings confirmed that DEHP induced a ROS-mediated imbalance in pyroptosis and mitophagy in immature rat testes via XBP1. Moreover, XBP1 might be the key target in DEHP-related testis dysfunction.
Collapse
Affiliation(s)
- Yifan Hong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiazhu Zhou
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qi Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jing Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yuexin Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Chunlan Long
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Lianju Shen
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiangqin Zheng
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Dinggang Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xia Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Chenjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| |
Collapse
|