1
|
Shih CC, Lin WL, Chuu CP, Lin C, Mi FL, Liu CW, Lu HY. Modified citrus pectin protects aortic dissection development involving macrophage pyroptosis. Arch Biochem Biophys 2025; 769:110428. [PMID: 40252790 DOI: 10.1016/j.abb.2025.110428] [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: 10/24/2024] [Revised: 03/28/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Uncontrolled aortic aneurysms can progress to aortic dissection (AD), a severe vascular disorder characterized by hematoma formation in the aortic wall, with inflammation playing a crucial role. Galectin-3 (Gal-3), a 26-kDa lectin, regulates many aspects of inflammatory cell behavior. Inhibition of Gal-3 ameliorates diabetic neuroinflammation and cardiomyopathy. Modified citrus pectin (MCP) is a PH-modified dietetic supplement produced from citrus pectin. This study investigates the therapeutic potential of MCP, which has a known affinity for Gal-3, in AD. METHODS A murine model of AD was induced by β-aminopropionitrile fumarate (BAPN)/angiotensin II (Ang-II) and treated orally with either 100 mg/kg MCP or vehicle. In vitro, H2O2 treatment was applied to RAW264.7 cells to detect macrophage death and pyroptosis. RESULTS MCP administration significantly reduced AD incidence and mortality, with decreased inflammatory cell infiltration in the aorta. MCP downregulated genes associated with inflammation and pyroptosis. In vitro, MCP mitigated macrophage death and pyroptosis induced by H2O2 treatment. The study suggests that MCP's protective effects are due to its interference with the Gal-3 and TLR4 interaction, inhibiting pyroptotic macrophage-induced inflammation. CONCLUSION MCP could improve patient outcomes and reduce progression to severe forms of AD. Clinically, MCP could serve as supportive therapy to prevent and delay aortic dissection, particularly during the acute stage of uncomplicated type B AD in patients with Marfan syndrome or abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Chun-Che Shih
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan
| | - Wei-Lun Lin
- Department of Medical Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 407219, Taiwan; Institute of Biomedical Sciences, MacKay Medical College, No. 46, Section 3, Zhongzheng Rd, New Taipei City, 252, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chi Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Fwu-Long Mi
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chen-Wei Liu
- Department of Basic Medical Science, College of Medicine, University of Arizona, Phoenix, AZ, 85721, USA
| | - Hsin-Ying Lu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan; Department of Physical Medicine and Rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
2
|
Fan X, Cheng Z, Shao R, Ye K, Chen X, Cai X, Dai S, Tang Z, Shi S, Zheng W, Huang W, Han J, Ye B. The novel GSDMD inhibitor GI-Y2 exerts antipyroptotic effects to reduce atherosclerosis. Clin Transl Med 2025; 15:e70263. [PMID: 40045452 PMCID: PMC11882392 DOI: 10.1002/ctm2.70263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
INTRODUCTION Gasdermin D (GSDMD) and the pyroptosis it mediates are importantly involved in cardiovascular diseases (CVDs). Identifying and developing new inhibitors of GSDMD could be a promising strategy for treating pyroptosis-mediated diseases, such as atherosclerosis. OBJECTIVES We aimed to develop new inhibitor of GSDMD in atherosclerosis, as well as clarify the mechanisms underlying this inhibiting effect. METHODS Surface plasmon resonance and pull-down assay were used to identify the amino acid sites of GSDMD inhibited by GI-Y2. A mouse model of atherosclerosis was established by feeding a high-fat diet for 12 weeks. After treating mice with GI-Y2 (10 or 20 mg/kg, i.g.), the lipid plaque area on the arterial intimal surface, lipid deposition, collagen deposition and pyroptosis levels in aortic root sections were evaluated. Additionally, further treatment of atherosclerotic mice with macrophage membrane-encapsulated GI-Y2 was conducted to enhance the targeting ability of GI-Y2 to atherosclerotic plaques. RESULTS In this study, we confirmed GI-Y2 as a novel inhibitor of GSDMD via structure-based virtual screening and pharmacological validation. Mechanistically, GI-Y2 directly interacts with the Arg10 residue of GSDMD and reduces the membrane binding of GSDMD-N. Functionally, we revealed that GI-Y2 inhibits the formation of atherosclerotic plaques by targeting GSDMD. Similarly, GI-Y2 reduces pyroptosis and macrophage infiltration in atherosclerosis. Furthermore, we constructed macrophage membrane-coated GI-Y2 nanoparticles to enhance the targeting of GI-Y2 to macrophages in atheromatous plaques and demonstrated its vascular protective effect in vivo. CONCLUSION This work demonstrated that GI-Y2 can potentially alleviate CVDs by targeting GSDMD and provided a new compound for the study of GSDMD-mediated pyroptosis. KEY POINTS We preliminarily confirmed GI-Y2 as a novel inhibitor of GSDMD via structure-based virtual screening and pharmacological validation. GI-Y2 directly interacts with GSDMD and reduces the membrane binding of GSDMD-N via the Arg10 residue. GI-Y2 inhibits the formation of atherosclerotic plaques by targeting GSDMD and GI-Y2 reduces pyroptosis and macrophage infiltration in atherosclerosis. We constructed macrophage membrane-coated GI-Y2 nanoparticles to enhance the targeting of GI-Y2 to macrophages in atheromatous plaques and demonstrated its vascular protective effect in vivo.
Collapse
Affiliation(s)
- Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhouthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
- Department of CardiologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingZhejiangChina
| | - Zhenfeng Cheng
- Huzhou Central HospitalAffiliated Central Hospital of Huzhou UniversityHuzhouChina
| | - Ruiyin Shao
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhouthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Keke Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhouthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xudong Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhouthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhouthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of WenzhouDepartment of EmergencyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Zhixuan Tang
- First School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
| | - Si Shi
- First School of MedicineWenzhou Medical UniversityWenzhouZhejiangChina
| | - Wenyuan Zheng
- Key Laboratory of Precision Medicine For Atherosclerosis Disease of Zhejiang ProvinceDepartment of CardiologyAffiliated First Hospital of Ningbo UniversityNingboZhejiangChina
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhouthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Jibo Han
- Department of CardiologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingZhejiangChina
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhouthe First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
3
|
He G, Ni Y, Hua R, Wan H, Tan Y, Chen Q, Xu S, Yang Y, Zhang L, Shu W, Huang KB, Mo Y, Liang H, Chen M. Latexin deficiency limits foam cell formation and ameliorates atherosclerosis by promoting macrophage phenotype differentiation. Cell Death Dis 2024; 15:754. [PMID: 39424784 PMCID: PMC11492231 DOI: 10.1038/s41419-024-07141-3] [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: 04/05/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Latexin (LXN) is abundant in macrophages and plays critical roles in inflammation. Much is known about macrophages in atherosclerosis, the role of macrophage LXN in atherosclerosis has remained elusive. Here, the expression of LXN in human and mouse atherosclerotic lesions was examined by immunofluorescence and immunohistochemistry. LXN knockout and LXN/ApoE double-knockout mice were generated to evaluate the functions of LXN in atherosclerosis. Bone marrow transplantation (BMT) experimentation was carried out to determine whether macrophage LXN regulates atherosclerosis. We found that LXN is enriched in human and murine atherosclerotic lesions, mainly localized to macrophages. LXN deletion ameliorated atherosclerosis in ApoE-/- mice. BMT demonstrate that deletion of LXN in bone marrow protects ApoE-/- mice against atherosclerosis. Mechanistically, we found that LXN targets and inhibits JAK1 in macrophages. LXN deficiency stimulates the JAK1/STAT3/ABC transporter pathway, thereby enhancing the anti-inflammatory and anti-oxidant phenotype, cholesterol efflux, subsequently minimizing foam cell formation and atherosclerosis. Gene therapy by treatment of atherosclerotic mice with adeno-associated virus harbouring LXN-depleting shRNA attenuated the disease phenotype. In summary, our study provides new clues for the role of LXN in the pathological regulation of atherosclerosis, and determines that LXN is a target for preventing atherosclerosis, which may be a potential new anti-atherosclerosis therapeutic target.
Collapse
Affiliation(s)
- Guozhang He
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yuanting Ni
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Rong Hua
- Department of Scientific Research, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huaibin Wan
- Heyuan Research Center for Cardiovascular Diseases, Department of Cardiology, the Fifth Affiliated Hospital of Jinan University, Heyuan, Guangdong, China
| | - Yanhui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Qiwei Chen
- Department of Scientific Research, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shaohua Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yuzhong Yang
- Department of Pathology, Affiliated Hospital of Guilin Medical College, Guilin, China
| | - Lijun Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Ke-Bin Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yi Mo
- Biobank department, The reproductive hospital of Guangxi Zhuang autonomous region, Nanning, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China.
| | - Ming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China.
| |
Collapse
|
4
|
Cheng CK, Yi M, Wang L, Huang Y. Role of gasdermin D in inflammatory diseases: from mechanism to therapeutics. Front Immunol 2024; 15:1456244. [PMID: 39253076 PMCID: PMC11381298 DOI: 10.3389/fimmu.2024.1456244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Inflammatory diseases compromise a clinically common and diverse group of conditions, causing detrimental effects on body functions. Gasdermins (GSDM) are pore-forming proteins, playing pivotal roles in modulating inflammation. Belonging to the GSDM family, gasdermin D (GSDMD) actively mediates the pathogenesis of inflammatory diseases by mechanistically regulating different forms of cell death, particularly pyroptosis, and cytokine release, in an inflammasome-dependent manner. Aberrant activation of GSDMD in different types of cells, such as immune cells, cardiovascular cells, pancreatic cells and hepatocytes, critically contributes to the persistent inflammation in different tissues and organs. The contributory role of GSDMD has been implicated in diabetes mellitus, liver diseases, cardiovascular diseases, neurodegenerative diseases, and inflammatory bowel disease (IBD). Clinically, alterations in GSDMD levels are potentially indicative to the occurrence and severity of diseases. GSDMD inhibition might represent an attractive therapeutic direction to counteract the progression of inflammatory diseases, whereas a number of GSDMD inhibitors have been shown to restrain GSDMD-mediated pyroptosis through different mechanisms. This review discusses the current understanding and future perspectives on the role of GSDMD in the development of inflammatory diseases, as well as the clinical insights of GSDMD alterations, and therapeutic potential of GSDMD inhibitors against inflammatory diseases. Further investigation on the comprehensive role of GSDM shall deepen our understanding towards inflammation, opening up more diagnostic and therapeutic opportunities against inflammatory diseases.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Min Yi
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Xiang Q, Geng ZX, Yi X, Wei X, Zhu XH, Jiang DS. PANoptosis: a novel target for cardiovascular diseases. Trends Pharmacol Sci 2024; 45:739-756. [PMID: 39003157 DOI: 10.1016/j.tips.2024.06.002] [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: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024]
Abstract
PANoptosis is a unique innate immune inflammatory lytic cell death pathway initiated by an innate immune sensor and driven by caspases and RIPKs. As a distinct pathway, the execution of PANoptosis cannot be hindered by targeting other cell death pathways, such as pyroptosis, apoptosis, or necroptosis. Instead, targeting key PANoptosome components can serve as a strategy to prevent this form of cell death. Given the physiological relevance in several diseases, PANoptosis is a pivotal therapeutic target. Notably, previous research has primarily focused on the role of PANoptosis in cancer and infectious and inflammatory diseases. By contrast, its role in cardiovascular diseases has not been comprehensively discussed. Here, we review the available evidence on PANoptosis in cardiovascular diseases, including cardiomyopathy, atherosclerosis, myocardial infarction, myocarditis, and aortic aneurysm and dissection, and explore a variety of agents that target PANoptosis, with the overarching goal of providing a novel complementary approach to combatting cardiovascular diseases.
Collapse
Affiliation(s)
- Qi Xiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen-Xi Geng
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xue-Hai Zhu
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
6
|
Jia Z, Zhang X, Li Z, Yan H, Tian X, Luo C, Ma K, Li L, Zhang L. Hydrogen sulfide mitigates ox‑LDL‑induced NLRP3/caspase‑1/GSDMD dependent macrophage pyroptosis by S‑sulfhydrating caspase‑1. Mol Med Rep 2024; 30:135. [PMID: 38873985 PMCID: PMC11188054 DOI: 10.3892/mmr.2024.13259] [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: 08/02/2023] [Accepted: 04/19/2024] [Indexed: 06/15/2024] Open
Abstract
Macrophage pyroptosis mediates vascular inflammation and atherosclerosis (AS). Hydrogen sulfide (H2S) exerts a protective role in preventing inflammation and AS. However, its molecular mechanisms of regulating the pyroptosis signaling pathway and inhibiting macrophage pyroptosis remain unexplored. The present study aimed to determine whether H2S mitigates macrophage pyroptosis by downregulating the pyroptosis signaling pathway and S‑sulfhydrating caspase‑1 under the stimulation of oxidized low‑density lipoprotein (ox‑LDL), a pro‑atherosclerotic factor. Macrophages derived from THP‑1 monocytes were pre‑treated using exogenous H2S donors sodium hydrosulfide (NaHS) and D,L‑propargylglycine (PAG), a pharmacological inhibitor of endogenous H2S‑producing enzymes, alone or in combination. Subsequently, cells were stimulated with ox‑LDL or the desulfhydration reagent dithiothreitol (DTT) in the presence or absence of NaHS and/or PAG. Following treatment, the levels of H2S in THP‑1 derived macrophages were measured by a methylene blue colorimetric assay. The pyroptotic phenotype of THP‑1 cells was observed and evaluated by light microscopy, Hoechst 33342/propidium iodide fluorescent staining and lactate dehydrogenase (LDH) release assay. Caspase‑1 activity in THP‑1 cells was assayed by caspase‑1 activity assay kit. Immunofluorescence staining was used to assess the accumulation of active caspase‑1. Western blotting and ELISA were performed to determine the expression of pyroptosis‑specific markers (NLRP3, pro‑caspase‑1, caspase‑1, GSDMD and GSDMD‑N) in cells and the secretion of pyroptosis‑related cytokines [interleukin (IL)‑1β and IL‑18] in the cell‑free media, respectively. The S‑sulfhydration of pro‑caspase‑1 in cells was assessed using a biotin switch assay. ox‑LDL significantly induced macrophage pyroptosis by activating the pyroptosis signaling pathway. Inhibition of endogenous H2S synthesis by PAG augmented the pro‑pyroptotic effects of ox‑LDL. Conversely, exogenous H2S (NaHS) ameliorated ox‑LDL‑and ox‑LDL + PAG‑induced macrophage pyroptosis by suppressing the activation of the pyroptosis signaling pathway. Mechanistically, ox‑LDL and the DTT increased caspase‑1 activity and downstream events (IL‑1β and IL‑18 secretion) of the caspase‑1‑dependent pyroptosis pathway by reducing S‑sulfhydration of pro‑caspase‑1. Conversely, NaHS increased S‑sulfhydration of pro‑caspase‑1, reducing caspase‑1 activity and caspase‑1‑dependent macrophage pyroptosis. The present study demonstrated the molecular mechanism by which H2S ameliorates macrophage pyroptosis by suppressing the pyroptosis signaling pathway and S‑sulfhydration of pro‑caspase‑1, thereby suppressing the generation of active caspase-1 and activity of caspase-1.
Collapse
Affiliation(s)
- Zhenli Jia
- Ministry of Education Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
| | - Xulin Zhang
- Department of Blood Transfusion, Shenzhen Children's Hospital, Shenzhen, Guangdong 518034, P.R. China
| | - Zhiyi Li
- Ministry of Education Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
| | - Hanyu Yan
- Ministry of Education Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
| | - Xiangqin Tian
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Chenghua Luo
- Ministry of Education Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
| | - Ketao Ma
- Ministry of Education Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Ling Li
- Department of Medical Morphology, Medical Teaching Experimental Center, School of Medicine, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Liang Zhang
- Ministry of Education Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| |
Collapse
|
7
|
Guo J, Zhang Q, Li Z, Qin M, Shi J, Wang Y, Ai W, Ju J, Samura M, Tsao PS, Xu B. Gasdermin D Inhibitor Necrosulfonamide Alleviates Angiotensin II-Induced Abdominal Aortic Aneurysms in Apolipoprotein E-Deficient Mice. Biomolecules 2024; 14:726. [PMID: 38927129 PMCID: PMC11201507 DOI: 10.3390/biom14060726] [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: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic aortic disease that lacks effective pharmacological therapies. This study was performed to determine the influence of treatment with the gasdermin D inhibitor necrosulfonamide on experimental AAAs. AAAs were induced in male apolipoprotein E-deficient mice by subcutaneous angiotensin II infusion (1000 ng/kg body weight/min), with daily administration of necrosulfonamide (5 mg/kg body weight) or vehicle starting 3 days prior to angiotensin II infusion for 30 days. Necrosulfonamide treatment remarkably suppressed AAA enlargement, as indicated by reduced suprarenal maximal external diameter and surface area, and lowered the incidence and reduced the severity of experimental AAAs. Histologically, necrosulfonamide treatment attenuated medial elastin breaks, smooth muscle cell depletion, and aortic wall collagen deposition. Macrophages, CD4+ T cells, CD8+ T cells, and neovessels were reduced in the aneurysmal aortas of necrosulfonamide- as compared to vehicle-treated angiotensin II-infused mice. Atherosclerosis and intimal macrophages were also substantially reduced in suprarenal aortas from angiotensin II-infused mice following necrosulfonamide treatment. Additionally, the levels of serum interleukin-1β and interleukin-18 were significantly lower in necrosulfonamide- than in vehicle-treated mice without affecting body weight gain, lipid levels, or blood pressure. Our findings indicate that necrosulfonamide reduced experimental AAAs by preserving aortic structural integrity as well as reducing mural leukocyte accumulation, neovessel formation, and systemic levels of interleukin-1β and interleukin-18. Thus, pharmacologically inhibiting gasdermin D activity may lead to the establishment of nonsurgical therapies for clinical AAA disease.
Collapse
Affiliation(s)
- Jia Guo
- Department of Cardiovascular Medicine, First Hospital Shanxi Medical University, Taiyuan 030001, Shanxi, China; (Q.Z.); (M.Q.); (J.S.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (W.A.); (J.J.); (M.S.); (B.X.)
| | - Qing Zhang
- Department of Cardiovascular Medicine, First Hospital Shanxi Medical University, Taiyuan 030001, Shanxi, China; (Q.Z.); (M.Q.); (J.S.)
| | - Zhidong Li
- Department of Pharmacology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Min Qin
- Department of Cardiovascular Medicine, First Hospital Shanxi Medical University, Taiyuan 030001, Shanxi, China; (Q.Z.); (M.Q.); (J.S.)
| | - Jinyun Shi
- Department of Cardiovascular Medicine, First Hospital Shanxi Medical University, Taiyuan 030001, Shanxi, China; (Q.Z.); (M.Q.); (J.S.)
| | - Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China;
| | - Wenjia Ai
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (W.A.); (J.J.); (M.S.); (B.X.)
| | - Junjie Ju
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (W.A.); (J.J.); (M.S.); (B.X.)
| | - Makoto Samura
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (W.A.); (J.J.); (M.S.); (B.X.)
| | - Philip S Tsao
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA;
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (W.A.); (J.J.); (M.S.); (B.X.)
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| |
Collapse
|
8
|
Fan X, Han J, Zhong L, Zheng W, Shao R, Zhang Y, Shi S, Lin S, Huang Z, Huang W, Cai X, Ye B. Macrophage-Derived GSDMD Plays an Essential Role in Atherosclerosis and Cross Talk Between Macrophages via the Mitochondria-STING-IRF3/NF-κB Axis. Arterioscler Thromb Vasc Biol 2024; 44:1365-1378. [PMID: 38695170 DOI: 10.1161/atvbaha.123.320612] [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: 12/19/2023] [Accepted: 04/16/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND Macrophages play a crucial role in atherosclerotic plaque formation, and the death of macrophages is a vital factor in determining the fate of atherosclerosis. GSDMD (gasdermin D)-mediated pyroptosis is a programmed cell death, characterized by membrane pore formation and inflammatory factor release. METHODS ApoE-/- and Gsdmd-/- ApoE-/- mice, bone marrow transplantation, and AAV (adeno-associated virus serotype 9)-F4/80-shGSDMD (shRNA-GSDMD) were used to examine the effect of macrophage-derived GSDMD on atherosclerosis. Single-cell RNA sequencing was used to investigate the changing profile of different cellular components and the cellular localization of GSDMD during atherosclerosis. RESULTS First, we found that GSDMD is activated in human and mouse atherosclerotic plaques and Gsdmd-/- attenuates the atherosclerotic lesion area in high-fat diet-fed ApoE-/- mice. We performed single-cell RNA sequencing of ApoE-/- and Gsdmd-/- ApoE-/- mouse aortas and showed that GSDMD is principally expressed in atherosclerotic macrophages. Using bone marrow transplantation and AAV-F4/80-shGSDMD, we identified the potential role of macrophage-derived GSDMD in aortic pyroptosis and atherosclerotic injuries in vivo. Mechanistically, GSDMD contributes to mitochondrial perforation and mitochondrial DNA leakage and subsequently activates the STING (stimulator of interferon gene)-IRF3 (interferon regulatory factor 3)/NF-κB (nuclear factor kappa B) axis. Meanwhile, GSDMD regulates the STING pathway activation and macrophage migration via cytokine secretion. Inhibition of GSDMD with GSDMD-specific inhibitor GI-Y1 (GSDMD inhibitor Y1) can effectively alleviate the progression of atherosclerosis. CONCLUSIONS Our study has provided a novel macrophage-derived GSDMD mechanism in the promotion of atherosclerosis and demonstrated that GSDMD can be a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China (J.H.)
| | - Lingfeng Zhong
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Wenyuan Zheng
- Key Laboratory of Precision Medicine for Atherosclerosis Disease of Zhejiang Province, Department of Cardiology, Affiliated First Hospital of Ningbo University, Zhejiang, China (W.Z., B.Y.)
| | - Ruiyin Shao
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Yucong Zhang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Si Shi
- First School of Medicine, Wenzhou Medical University, Zhejiang, China (S.S.)
| | - Shuang Lin
- Department of Cardiology, Ningbo Medical Center Li Huili Hospital, Zhejiang, China (S.L.)
| | - Zhouqing Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
- Key Laboratory of Precision Medicine for Atherosclerosis Disease of Zhejiang Province, Department of Cardiology, Affiliated First Hospital of Ningbo University, Zhejiang, China (W.Z., B.Y.)
| |
Collapse
|
9
|
De Meyer GRY, Zurek M, Puylaert P, Martinet W. Programmed death of macrophages in atherosclerosis: mechanisms and therapeutic targets. Nat Rev Cardiol 2024; 21:312-325. [PMID: 38163815 DOI: 10.1038/s41569-023-00957-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a progressive inflammatory disorder of the arterial vessel wall characterized by substantial infiltration of macrophages, which exert both favourable and detrimental functions. Early in atherogenesis, macrophages can clear cytotoxic lipoproteins and dead cells, preventing cytotoxicity. Efferocytosis - the efficient clearance of dead cells by macrophages - is crucial for preventing secondary necrosis and stimulating the release of anti-inflammatory cytokines. In addition, macrophages can promote tissue repair and proliferation of vascular smooth muscle cells, thereby increasing plaque stability. However, advanced atherosclerotic plaques contain large numbers of pro-inflammatory macrophages that secrete matrix-degrading enzymes, induce death in surrounding cells and contribute to plaque destabilization and rupture. Importantly, macrophages in the plaque can undergo apoptosis and several forms of regulated necrosis, including necroptosis, pyroptosis and ferroptosis. Regulated necrosis has an important role in the formation and expansion of the necrotic core during plaque progression, and several triggers for necrosis are present within atherosclerotic plaques. This Review focuses on the various forms of programmed macrophage death in atherosclerosis and the pharmacological interventions that target them as a potential means of stabilizing vulnerable plaques and improving the efficacy of currently available anti-atherosclerotic therapies.
Collapse
Affiliation(s)
- Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Michelle Zurek
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
10
|
Traughber CA, Timinski K, Prince A, Bhandari N, Neupane K, Khan MR, Opoku E, Opoku E, Brubaker G, Shin J, Hong J, Kanuri B, Ertugral EG, Nagareddy PR, Kothapalli CR, Cherepanova O, Smith JD, Gulshan K. Disulfiram Reduces Atherosclerosis and Enhances Efferocytosis, Autophagy, and Atheroprotective Gut Microbiota in Hyperlipidemic Mice. J Am Heart Assoc 2024; 13:e033881. [PMID: 38563369 PMCID: PMC11262521 DOI: 10.1161/jaha.123.033881] [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: 03/06/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Pyroptosis executor GsdmD (gasdermin D) promotes atherosclerosis in mice and humans. Disulfiram was recently shown to potently inhibit GsdmD, but the in vivo efficacy and mechanism of disulfiram's antiatherosclerotic activity is yet to be explored. METHODS AND RESULTS We used human/mouse macrophages, endothelial cells, and smooth muscle cells and a hyperlipidemic mouse model of atherosclerosis to determine disulfiram antiatherosclerotic efficacy and mechanism. The effects of disulfiram on several atheroprotective pathways such as autophagy, efferocytosis, phagocytosis, and gut microbiota were determined. Atomic force microscopy was used to determine the effects of disulfiram on the biophysical properties of the plasma membrane of macrophages. Disulfiram-fed hyperlipidemic apolipoprotein E-/- mice showed significantly reduced interleukin-1β release upon in vivo Nlrp3 (NLR family pyrin domain containing 3) inflammasome activation. Disulfiram-fed mice showed smaller atherosclerotic lesions (~27% and 29% reduction in males and females, respectively) and necrotic core areas (~50% and 46% reduction in males and females, respectively). Disulfiram induced autophagy in macrophages, smooth muscle cells, endothelial cells, hepatocytes/liver, and atherosclerotic plaques. Disulfiram modulated other atheroprotective pathways (eg, efferocytosis, phagocytosis) and gut microbiota. Disulfiram-treated macrophages showed enhanced phagocytosis/efferocytosis, with the mechanism being a marked increase in cell-surface expression of efferocytic receptor MerTK. Atomic force microscopy analysis revealed altered biophysical properties of disulfiram-treated macrophages, showing increased order-state of plasma membrane and increased adhesion strength. Furthermore, 16sRNA sequencing of disulfiram-fed hyperlipidemic mice showed highly significant enrichment in atheroprotective gut microbiota Akkermansia and a reduction in atherogenic Romboutsia species. CONCLUSIONS Taken together, our data show that disulfiram can simultaneously modulate several atheroprotective pathways in a GsdmD-dependent as well as GsdmD-independent manner.
Collapse
Affiliation(s)
- C. Alicia Traughber
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Kara Timinski
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Ashutosh Prince
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Nilam Bhandari
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Kalash Neupane
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Mariam R. Khan
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Esther Opoku
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Emmanuel Opoku
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Gregory Brubaker
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Junchul Shin
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Babunageswararao Kanuri
- Department of Internal Medicine, Cardiovascular SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Elif G. Ertugral
- Department of Chemical & Biomedical EngineeringCleveland State UniversityClevelandOHUSA
| | - Prabhakara R. Nagareddy
- Department of Internal Medicine, Cardiovascular SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | | | - Olga Cherepanova
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Jonathan D. Smith
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Kailash Gulshan
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| |
Collapse
|
11
|
Huang B, Zou Z, Li Y, Chen H, Lai K, Yuan Y, Xu Y. Gasdermin D-Mediated Pyroptosis Promotes the Development of Atherosclerosis. J Transl Med 2024; 104:100337. [PMID: 38266921 DOI: 10.1016/j.labinv.2024.100337] [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: 05/20/2023] [Revised: 12/16/2023] [Accepted: 01/16/2024] [Indexed: 01/26/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory cardiovascular disease with a high-morbidity and mortality rate. An increasing number of studies have addressed the crucial contribution of gasdermin D (GSDMD)-mediated pyroptosis, which is triggered by the inflammasomes to the development of atherosclerosis. However, the underlying mechanism is still unclear. This study aimed to uncover the detailed role of GSDMD in the development of atherosclerosis. An atherosclerotic model was established in Gsdmd-/-/Ldlr-/- mice and Gsdmd+/+/Ldlr-/- mice fed with a high-fat diet. The atherosclerotic lesions, the activation of GSDMD, and the expression level of inflammatory cytokines and chemokines were evaluated. Gsdmd deletion ameliorated the atherosclerotic lesion sizes and the infiltration of immune cells and inflammatory cells in the aortas of mice. Additionally, Gsdmd deletion suppressed the pyroptosis of macrophages and endothelial cells induced by the serum of Ldlr-/- mice fed with a high-fat diet. Furthermore, the formation of neutrophil extracellular traps was also attenuated by knockout of Gsdmd. Bone marrow chimeras confirmed that the genetic deficiency of Gsdmd in both immune cells and intrinsic cells played a role in the promotion of arteriosclerosis. Collectively, our study demonstrated that Gsdmd deletion hindered the pathogenesis of atherosclerosis by inhibiting endothelial cell and macrophage cell death, and the formation of neutrophil extracellular traps.
Collapse
Affiliation(s)
- Bangbang Huang
- Department of Geriatrics, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhenhuan Zou
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Nephrology, Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yinshuang Li
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Nephrology, Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hui Chen
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Kunmei Lai
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Nephrology, Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ying Yuan
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Nephrology, Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yanfang Xu
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Nephrology, Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
12
|
Makuch M, Stepanechko M, Bzowska M. The dance of macrophage death: the interplay between the inevitable and the microenvironment. Front Immunol 2024; 15:1330461. [PMID: 38576612 PMCID: PMC10993711 DOI: 10.3389/fimmu.2024.1330461] [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: 10/30/2023] [Accepted: 02/26/2024] [Indexed: 04/06/2024] Open
Abstract
Macrophages are highly plastic cells ubiquitous in various tissues, where they perform diverse functions. They participate in the response to pathogen invasion and inflammation resolution following the immune response, as well as the maintenance of homeostasis and proper tissue functions. Macrophages are generally considered long-lived cells with relatively strong resistance to numerous cytotoxic factors. On the other hand, their death seems to be one of the principal mechanisms by which macrophages perform their physiological functions or can contribute to the development of certain diseases. In this review, we scrutinize three distinct pro-inflammatory programmed cell death pathways - pyroptosis, necroptosis, and ferroptosis - occurring in macrophages under specific circumstances, and explain how these cells appear to undergo dynamic yet not always final changes before ultimately dying. We achieve that by examining the interconnectivity of these cell death types, which in macrophages seem to create a coordinated and flexible system responding to the microenvironment. Finally, we discuss the complexity and consequences of pyroptotic, necroptotic, and ferroptotic pathway induction in macrophages under two pathological conditions - atherosclerosis and cancer. We summarize damage-associated molecular patterns (DAMPs) along with other microenvironmental factors, macrophage polarization states, associated mechanisms as well as general outcomes, as such a comprehensive look at these correlations may point out the proper methodologies and potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Małgorzata Bzowska
- Department of Immunology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
13
|
Lawlor KE, Murphy JM, Vince JE. Gasdermin and MLKL necrotic cell death effectors: Signaling and diseases. Immunity 2024; 57:429-445. [PMID: 38479360 DOI: 10.1016/j.immuni.2024.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/29/2023] [Accepted: 02/14/2024] [Indexed: 01/22/2025]
Abstract
Diverse inflammatory conditions, from infections to autoimmune disease, are often associated with cellular damage and death. Apoptotic cell death has evolved to minimize its inflammatory potential. By contrast, necrotic cell death via necroptosis and pyroptosis-driven by membrane-damaging MLKL and gasdermins, respectively-can both initiate and propagate inflammatory responses. In this review, we provide insights into the function and regulation of MLKL and gasdermin necrotic effector proteins and drivers of plasma membrane rupture. We evaluate genetic evidence that MLKL- and gasdermin-driven necrosis may either provide protection against, or contribute to, disease states in a context-dependent manner. These cumulative insights using gene-targeted mice underscore the necessity for future research examining pyroptotic and necroptotic cell death in human tissue, as a basis for developing specific necrotic inhibitors with the potential to benefit a spectrum of pathological conditions.
Collapse
Affiliation(s)
- Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - James M Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
14
|
Mulla J, Katti R, Scott MJ. The Role of Gasdermin-D-Mediated Pyroptosis in Organ Injury and Its Therapeutic Implications. Organogenesis 2023; 19:2177484. [PMID: 36967609 PMCID: PMC9980590 DOI: 10.1080/15476278.2023.2177484] [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: 09/15/2022] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
Gasdermin-D (GSDMD) belongs to the Gasdermin family (GSDM), which are pore-forming effector proteins that facilitate inflammatory cell death, also known as pyroptosis. This type of programmed cell death is dependent on inflammatory caspase activation, which cleaves gasdermin-D (GSDMD) to form membrane pores and initiates the release of pro-inflammatory cytokines. Pyroptosis plays an important role in achieving immune regulation and homeostasis within various organ systems. The role of GSDMD in pyroptosis has been extensively studied in recent years. In this review, we summarize the role of GSDMD in cellular and organ injury mediated by pyroptosis. We will also provide an outlook on GSDMD therapeutic targets in various organ systems.
Collapse
Affiliation(s)
- Joud Mulla
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rohan Katti
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melanie J. Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Traughber CA, Timinski K, Prince A, Bhandari N, Neupane K, Khan MR, Opoku E, Opoku E, Brubaker G, Nageshwar K, Ertugral EG, Naggareddy P, Kothapalli CR, Smith JD, Gulshan K. Disulfiram reduces atherosclerosis and enhances efferocytosis, autophagy, and atheroprotective gut microbiota in hyperlipidemic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562757. [PMID: 37905037 PMCID: PMC10614849 DOI: 10.1101/2023.10.17.562757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Pyroptosis executor Gasdermin (GsdmD) promotes atherosclerosis in mice and humans. Disulfiram (DSF) was recently shown to potently inhibit GsdmD, but the in-vivo efficacy and mechanism of DSF's anti-atherosclerotic activity is yet to be explored. We used human/mouse macrophages and a hyperlipidemic mouse model of atherosclerosis to determine DSF anti-atherosclerotic efficacy and mechanism. DSF-fed hyperlipidemic apoE -/- mice showed significantly reduced IL-1β release upon in-vivo Nlrp3 inflammasome assembly and showed smaller atherosclerotic lesions (∼27% and 29% reduction in males and females, respectively). The necrotic core area was also smaller (∼50% and 46% reduction in DSF-fed males and females, respectively). DSF induced autophagy in macrophages, hepatocytes/liver, and in atherosclerotic plaques. DSF modulated other atheroprotective pathways such as efferocytosis, phagocytosis, and gut microbiota. DSF-treated macrophages showed enhanced phagocytosis/efferocytosis, with a mechanism being a marked increase in cell-surface expression of efferocytic receptor MerTK. Atomic-force microscopy analysis revealed altered biophysical membrane properties of DSF treated macrophages, showing increased ordered-state of the plasma membrane and increased adhesion strength. Furthermore, the 16sRNA sequencing of DSF-fed hyperlipidemic mice showed highly significant enrichment in atheroprotective gut microbiota Akkermansia and a reduction in atherogenic Romboutsia species. Taken together, our data shows that DSF can simultaneously modulate multiple atheroprotective pathways, and thus may serve as novel adjuvant therapeutic to treat atherosclerosis.
Collapse
|
16
|
Bu LL, Yuan HH, Xie LL, Guo MH, Liao DF, Zheng XL. New Dawn for Atherosclerosis: Vascular Endothelial Cell Senescence and Death. Int J Mol Sci 2023; 24:15160. [PMID: 37894840 PMCID: PMC10606899 DOI: 10.3390/ijms242015160] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Endothelial cells (ECs) form the inner linings of blood vessels, and are directly exposed to endogenous hazard signals and metabolites in the circulatory system. The senescence and death of ECs are not only adverse outcomes, but also causal contributors to endothelial dysfunction, an early risk marker of atherosclerosis. The pathophysiological process of EC senescence involves both structural and functional changes and has been linked to various factors, including oxidative stress, dysregulated cell cycle, hyperuricemia, vascular inflammation, and aberrant metabolite sensing and signaling. Multiple forms of EC death have been documented in atherosclerosis, including autophagic cell death, apoptosis, pyroptosis, NETosis, necroptosis, and ferroptosis. Despite this, the molecular mechanisms underlying EC senescence or death in atherogenesis are not fully understood. To provide a comprehensive update on the subject, this review examines the historic and latest findings on the molecular mechanisms and functional alterations associated with EC senescence and death in different stages of atherosclerosis.
Collapse
Affiliation(s)
- Lan-Lan Bu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.-L.B.); (D.-F.L.)
| | - Huan-Huan Yuan
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
| | - Ling-Li Xie
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Min-Hua Guo
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
| | - Duan-Fang Liao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.-L.B.); (D.-F.L.)
| | - Xi-Long Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
17
|
Martins-Marques T, Coutinho G, Kiss A. Editorial of the Special Issue: Cellular Mechanisms of Cardiovascular Disease. Biomedicines 2023; 11:2494. [PMID: 37760934 PMCID: PMC10525463 DOI: 10.3390/biomedicines11092494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular diseases (CVD) remain the major cause of mortality and disability worldwide, having contributed to 19 [...].
Collapse
Affiliation(s)
- Tânia Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Gonçalo Coutinho
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
- University Hospital and Center of Coimbra, Cardiothoracic Surgery Department, 3000-075 Coimbra, Portugal
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research, Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
18
|
Yarovinsky TO, Su M, Chen C, Xiang Y, Tang WH, Hwa J. Pyroptosis in cardiovascular diseases: Pumping gasdermin on the fire. Semin Immunol 2023; 69:101809. [PMID: 37478801 PMCID: PMC10528349 DOI: 10.1016/j.smim.2023.101809] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/13/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023]
Abstract
Pyroptosis is a form of programmed cell death associated with activation of inflammasomes and inflammatory caspases, proteolytic cleavage of gasdermin proteins (forming pores in the plasma membrane), and selective release of proinflammatory mediators. Induction of pyroptosis results in amplification of inflammation, contributing to the pathogenesis of chronic cardiovascular diseases such as atherosclerosis and diabetic cardiomyopathy, and acute cardiovascular events, such as thrombosis and myocardial infarction. While engagement of pyroptosis during sepsis-induced cardiomyopathy and septic shock is expected and well documented, we are just beginning to understand pyroptosis involvement in the pathogenesis of cardiovascular diseases with less defined inflammatory components, such as atrial fibrillation. Due to the danger that pyroptosis represents to cells within the cardiovascular system and the whole organism, multiple levels of pyroptosis regulation have evolved. Those include regulation of inflammasome priming, post-translational modifications of gasdermins, and cellular mechanisms for pore removal. While pyroptosis in macrophages is well characterized as a dramatic pro-inflammatory process, pyroptosis in other cell types within the cardiovascular system displays variable pathways and consequences. Furthermore, different cells and organs engage in local and distant crosstalk and exchange of pyroptosis triggers (oxidized mitochondrial DNA), mediators (IL-1β, S100A8/A9) and antagonists (IL-9). Development of genetic tools, such as Gasdermin D knockout animals, and small molecule inhibitors of pyroptosis will not only help us fully understand the role of pyroptosis in cardiovascular diseases but may result in novel therapeutic approaches inhibiting inflammation and progression of chronic cardiovascular diseases to reduce morbidity and mortality from acute cardiovascular events.
Collapse
Affiliation(s)
- Timur O Yarovinsky
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Meiling Su
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China
| | - Chaofei Chen
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China
| | - Yaozu Xiang
- Shanghai East Hospital, Key Laboratory of Arrhythmias of the Ministry of Education of China, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Kowloon, the Hong Kong Special Administrative Region of China
| | - John Hwa
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
19
|
Fang Z, Wu G, Sheng J, Ye B, Huang Z, Xu J, Zhang J, Han J, Han B, Xu J. Gasdermin D affects aortic vascular smooth muscle cell pyroptosis and Ang II-induced vascular remodeling. Heliyon 2023; 9:e16619. [PMID: 37303505 PMCID: PMC10248119 DOI: 10.1016/j.heliyon.2023.e16619] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are primarily responsible for vasoconstriction and the regulation of blood pressure1. Pyroptosis, a particular form of regulated cell death, is involved in multiple vascular injuries, including hypertensive vascular dysfunction. This pyroptotic cell death is mediated by the pore-forming protein of Gasdermin D (GSDMD). This study was designed to examine the direct effect of GSDMD on smooth muscle cell pyroptosis and vascular remodeling. Findings revealed that GSDMD was activated in Angiotensin (Ang) II- treated aortas. We then showed that genetic deletion of Gsdmd reduced vascular remodeling and aorta pyroptosis induced by Ang II in vivo. Aberrant expression of GSDMD by recombinant AAV9 virus carrying Gsdmd cDNA aggravated the level of pyroptosis in aortas of Ang II mice. Gain- and loss-of- function analysis further confirmed that GSDMD regulated the pyroptosis of murine aortic vascular smooth muscle cells (MOVAS) in an in vitro model of tumor necrosis factor (TNF)-α treatment, which was achieved by transfecting expressing plasmid or siRNA, respectively. Overall, this study provided evidence supporting the active involvement of GSDMD in smooth muscle cell pyroptosis and Ang II-induced mice vascular injury. This finding lends credence to GSDMD as a potential therapeutic target for hypertensive vascular remodeling via inhibiting pyroptosis.
Collapse
Affiliation(s)
- Zimin Fang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Sheng
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouqing Huang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianjiang Xu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jianqin Zhang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jibo Han
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bingjiang Han
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jiajun Xu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
20
|
Guo J, Shi J, Qin M, Wang Y, Li Z, Shoji T, Ikezoe T, Ge Y, Xu B. Pharmacological Inhibition of Gasdermin D Suppresses Angiotensin II-Induced Experimental Abdominal Aortic Aneurysms. Biomolecules 2023; 13:899. [PMID: 37371479 PMCID: PMC10295961 DOI: 10.3390/biom13060899] [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: 04/10/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Gasdermin D, a molecule downstream of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing inflammasome, forms the membrane pore for the secretion of interleukin (IL)-1β and IL-18, and also mediates pyroptosis. This study was to explore the influence of treatment with disulfiram, a small molecule inhibitor to gasdermin D, on the formation and progression of experimental abdominal aortic aneurysms (AAA). METHODS AAAs were induced in 10-week-old male apolipoprotein E deficient mice by subcutaneous infusion of angiotensin II (1000 ng/min/kg body weight) for 28 days via osmotic minipumps. Three days prior to angiotensin II infusion, disulfiram (50 mg/kg) or an equal volume of saline as the vehicle control was administered daily via oral gavage. The influence on experimental AAAs was analyzed by serial measurements of aortic diameters via ultrasonography, grading AAA severity and histopathology at sacrifice. Serum IL-1β and IL-18 levels, systolic blood pressure, total cholesterol, and triglyceride were also measured. Additional experiments assayed the influences on the cell viability and IL-1β secretion of in vitro activated macrophages. RESULTS Disulfiram significantly reduced the enlargement, incidence, and severity of angiotensin II-induced experimental AAAs with attenuation of medial elastin breaks, mural macrophage accumulation, and systolic blood pressure. The AAA suppression was also associated with reduced systemic levels of IL-1β but not IL-18. However, disulfiram treatment had no impact on body weight gain and lipid levels in aneurysmal mice. Additionally, disulfiram treatment also markedly reduced the secretion of IL-1β from activated macrophages with a limited effect on cell viability in vitro. CONCLUSIONS Gasdermin D inhibition by disulfiram attenuated angiotensin II-induced experimental AAAs with reduced systemic IL-1β levels and in vitro activated macrophage IL-1β secretion. Our study suggests that pharmacological gasdermin D inhibition may have translational potential for limiting clinical AAA progression.
Collapse
Affiliation(s)
- Jia Guo
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030001, China; (J.S.); (M.Q.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| | - Jinyun Shi
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030001, China; (J.S.); (M.Q.)
| | - Min Qin
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030001, China; (J.S.); (M.Q.)
| | - Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China;
| | - Zhidong Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China;
| | - Takahiro Shoji
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| | - Toru Ikezoe
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| | - Yingbin Ge
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China;
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.S.); (T.I.); (B.X.)
| |
Collapse
|
21
|
Abstract
The CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcome Study) and colchicine trials suggest an important role of inflammasomes and their major product IL-1β (interleukin 1β) in human atherosclerotic cardiovascular disease. Moreover, studies in mouse models indicate a causal role of inflammasomes and IL-1β in atherosclerosis. However, recent studies have led to a more granular view of the role of inflammasomes in atherosclerosis. Studies in hyperlipidemic mouse models suggest that prominent activation of the NLRP3 inflammasome requires a second hit such as defective cholesterol efflux, defective DNA repair, clonal hematopoiesis or diabetes. Similarly in humans some mutations promoting clonal hematopoiesis increase coronary artery disease risk in part by promoting inflammasome activation. Recent studies in mice and humans point to a wider role of the AIM2 (absent in melanoma 2) inflammasome in promoting cardiovascular disease including in some forms of clonal hematopoiesis and diabetes. These developments suggest a precision medicine approach in which treatments targeting inflammasomes or IL-1β might be best employed in clinical settings involving increased inflammasome activation.
Collapse
Affiliation(s)
- Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York (A.R.T.)
| | - Karin E Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington, Seattle (K.E.B.)
| |
Collapse
|
22
|
Traughber CA, Iacano AJ, Neupane K, Khan MR, Opoku E, Nunn T, Prince A, Sangwan N, Hazen SL, Smith JD, Gulshan K. Impavido attenuates inflammation, reduces atherosclerosis, and alters gut microbiota in hyperlipidemic mice. iScience 2023; 26:106453. [PMID: 37020959 PMCID: PMC10067757 DOI: 10.1016/j.isci.2023.106453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/14/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Impavido (Miltefosine) is an FDA-approved drug for treating leishmaniasis and primary amebic meningoencephalitis. We have shown previously that Miltefosine increased cholesterol release and dampened Nlrp3 inflammasome assembly in macrophages. Here, we show that Miltefosine reduced LPS-induced choline uptake by macrophages, and attenuated Nlrp3 inflammasome assembly in mice. Miltefosine-fed mice showed reduced plasma IL-1β in a polymicrobial cecal slurry model of systemic inflammation. Miltefosine-fed mice showed increased reverse cholesterol transport to the plasma, liver, and feces. Hyperlipidemic apoE-/- mice fed with WTD + Miltefosine showed significantly reduced weight gain and markedly reduced atherosclerotic lesions versus mice fed with WTD. The 16S rDNA sequencing and analysis of gut microbiota showed marked alterations in the microbiota profile of Miltefosine-fed hyperlipidemic apoE-/- versus control, with the most notable changes in Romboutsia and Bacteriodes species. Taken together, these data indicate that Miltefosine causes pleiotropic effects on lipid metabolism, inflammasome activity, atherosclerosis, and the gut microbiota.
Collapse
|
23
|
Traughber CA, Deshpande GM, Neupane K, Bhandari N, Khan MR, McMullen MR, Swaidani S, Opoku E, Muppala S, Smith JD, Nagy LE, Gulshan K. Myeloid-cell-specific role of Gasdermin D in promoting lung cancer progression in mice. iScience 2023; 26:106076. [PMID: 36844454 PMCID: PMC9947301 DOI: 10.1016/j.isci.2023.106076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/29/2022] [Accepted: 01/24/2023] [Indexed: 02/01/2023] Open
Abstract
The activities of the NLRP3 and AIM2 inflammasomes and Gasdermin D (GsdmD) are implicated in lung cancer pathophysiology but it's not clear if their contributions promote or retard lung cancer progression. Using a metastatic Lewis lung carcinoma (LLC) cell model, we show that GsdmD knockout (GsdmD-/-) mice form significantly fewer cancer foci in lungs, exhibit markedly decreased lung cancer metastasis, and show a significant ∼50% increase in median survival rate. The cleaved forms of GsdmD and IL-1β were detected in lung tumor tissue, indicating inflammasome activity in lung tumor microenvironment (TME). Increased migration and growth of LLC cells was observed upon exposure to the conditioned media derived from inflammasome-induced wild type, but not the GsdmD-/-, macrophages. Using bone marrow transplantations, we show a myeloid-specific contribution of GsdmD in lung cancer metastasis. Taken together, our data show that GsdmD plays a myeloid-specific role in lung cancer progression.
Collapse
Affiliation(s)
- C. Alicia Traughber
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH 44115, USA,Department of Biology, Geology, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA,Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gauravi M. Deshpande
- Digital Imaging Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kalash Neupane
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH 44115, USA,Department of Biology, Geology, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Nilam Bhandari
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH 44115, USA,Department of Biology, Geology, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Mariam R. Khan
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH 44115, USA,Department of Biology, Geology, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Megan R. McMullen
- Departments of Inflammation and Immunity and Gastroenterology/Hepatology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shadi Swaidani
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Emmanuel Opoku
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Santoshi Muppala
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jonathan D. Smith
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Laura E. Nagy
- Departments of Inflammation and Immunity and Gastroenterology/Hepatology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Northern Ohio Alcohol Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kailash Gulshan
- Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH 44115, USA,Department of Biology, Geology, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA,Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Corresponding author
| |
Collapse
|
24
|
Weng Y, Ye B, Lin J, Lin S, Zhong L, Huang W, Cai X, Wang W. Elevated circulating levels of gasdermin D are related to acute myocardial infarction and pyrogptosis. BMC Cardiovasc Disord 2022; 22:554. [PMID: 36544106 PMCID: PMC9769051 DOI: 10.1186/s12872-022-02998-8] [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: 06/16/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is one of the leading contributors to morbidity and mortality worldwide, with a prevalence of nearly three million people, and more than one million deaths reported in the United States every year. Gasdermin D (GSDMD) is involved in the development of atherosclerosis as a key protein of proptosis. This study was designed to determine the potential relationship of GSDMD with AMI in Chinese patients. METHODS One hundred patients with AMI and 50 controls were consecutively enrolled in this prospective observational study. GSDMD expression levels and other clinical variables in peripheral blood mononuclear cells (PBMCs) were measured upon admission to the hospital. All patients were followed up for 360 days, and the endpoint was considered the occurrence of major adverse cardiovascular events (MACE). RESULTS GSDMD expression levels in the PBMCs of patients with AMI were significantly higher than those in the controls. Moreover, our analysis showed that GSDMD was an independent biomarker of AMI and had a promising diagnostic ability for it. Finally, the results suggested that high expression of GSDMD and diabetes increased the risk of MACE after AMI. CONCLUSIONS This study indicated that the GSDMD expression level in PBMCs was elevated in AMI patients and was closely associated with the pyroptosis of AMI.
Collapse
Affiliation(s)
- Yawen Weng
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China ,grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, 200127 People’s Republic of China
| | - Bozhi Ye
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China
| | - Jiahui Lin
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China
| | - Shuang Lin
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China
| | - Lingfeng Zhong
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China
| | - Weijian Huang
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China
| | - Xueli Cai
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China
| | - Weiqi Wang
- grid.414906.e0000 0004 1808 0918Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, 325000 Zhejiang People’s Republic of China
| |
Collapse
|
25
|
Xu XD, Chen JX, Zhu L, Xu ST, Jiang J, Ren K. The emerging role of pyroptosis-related inflammasome pathway in atherosclerosis. Mol Med 2022; 28:160. [PMID: 36544112 PMCID: PMC9773468 DOI: 10.1186/s10020-022-00594-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis (AS), a chronic sterile inflammatory disorder, is one of the leading causes of mortality worldwide. The dysfunction and unnatural death of plaque cells, including vascular endothelial cells (VEC), macrophages, and vascular smooth muscle cells (VSMC), are crucial factors in the progression of AS. Pyroptosis was described as a form of cell death at least two decades ago. It is featured by plasma membrane swelling and rupture, cell lysis, and consequent robust release of cytosolic contents and pro-inflammatory mediators, including interleukin-1β (IL-1β), IL-18, and high mobility group box 1 (HMGB1). Pyroptosis of plaque cells is commonly observed in the initiation and development of AS, and the levels of pyroptosis-related proteins are positively correlated with plaque instability, indicating the crucial contribution of pyroptosis to atherogenesis. Furthermore, studies have also identified some candidate anti-atherogenic agents targeting plaque cell pyroptosis. Herein, we summarize the research progress in understating (1) the discovery and definition of pyroptosis; (2) the characterization and molecular mechanisms of pyroptosis; (3) the regulatory mechanisms of pyroptosis in VEC, macrophage, and VSMC, as well as their potential role in AS progression, aimed at providing therapeutic targets for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Xiao-Dan Xu
- grid.412679.f0000 0004 1771 3402Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui People’s Republic of China
| | - Jia-Xian Chen
- grid.443397.e0000 0004 0368 7493Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100 Hainan People’s Republic of China
| | - Lin Zhu
- grid.252251.30000 0004 1757 8247College of Nursing, Anhui University of Chinese Medicine, Hefei, 230012 Anhui People’s Republic of China
| | - Shu-Ting Xu
- grid.411971.b0000 0000 9558 1426Department of Nephrology, The Affiliated Hospital of Dalian Medical University, Dalian, 116044 Liaoning People’s Republic of China
| | - Jian Jiang
- grid.443397.e0000 0004 0368 7493Department of Organ Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100 Hainan People’s Republic of China
| | - Kun Ren
- grid.252251.30000 0004 1757 8247College of Nursing, Anhui University of Chinese Medicine, Hefei, 230012 Anhui People’s Republic of China ,grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100 Hainan People’s Republic of China
| |
Collapse
|
26
|
Puylaert P, Zurek M, Rayner KJ, De Meyer GRY, Martinet W. Regulated Necrosis in Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:1283-1306. [PMID: 36134566 DOI: 10.1161/atvbaha.122.318177] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During atherosclerosis, lipid-rich plaques are formed in large- and medium-sized arteries, which can reduce blood flow to tissues. This situation becomes particularly precarious when a plaque develops an unstable phenotype and becomes prone to rupture. Despite advances in identifying and treating vulnerable plaques, the mortality rate and disability caused by such lesions remains the number one health threat in developed countries. Vulnerable, unstable plaques are characterized by a large necrotic core, implying a prominent role for necrotic cell death in atherosclerosis and plaque destabilization. Necrosis can occur accidentally or can be induced by tightly regulated pathways. Over the past decades, different forms of regulated necrosis, including necroptosis, ferroptosis, pyroptosis, and secondary necrosis, have been identified, and these may play an important role during atherogenesis. In this review, we describe several forms of necrosis that may occur in atherosclerosis and how pharmacological modulation of these pathways can stabilize vulnerable plaques. Moreover, some challenges of targeting necrosis in atherosclerosis such as the presence of multiple death-inducing stimuli in plaques and extensive cross-talk between necrosis pathways are discussed. A better understanding of the role of (regulated) necrosis in atherosclerosis and the mechanisms contributing to plaque destabilization may open doors to novel pharmacological strategies and will enable clinicians to tackle the residual cardiovascular risk that remains in many atherosclerosis patients.
Collapse
Affiliation(s)
- Pauline Puylaert
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Michelle Zurek
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology and Immunology and Centre for Infection, Immunity and Inflammation, Faculty of Medicine, University of Ottawa, ON, Canada (K.J.R.).,University of Ottawa Heart Institute, ON, Canada (K.J.R.)
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Wim Martinet
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| |
Collapse
|