1
|
Han Y, Wu X, Shi X, Zhang G, Wang X, Wang C, Zhou H. AKAP1-STABILIZED TIMP-4 ATTENUATES ANG-II-INDUCED OXIDATIVE STRESS AND INFLAMMATION IN VASCULAR SMOOTH MUSCLE CELLS BY INACTIVATING THE NF-ΚB SIGNALING. Shock 2025; 63:750-759. [PMID: 39965635 DOI: 10.1097/shk.0000000000002557] [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: 02/20/2025]
Abstract
ABSTRACT Background: Oxidative stress and inflammation are key factors contributing to the complex pathogenesis of abdominal aortic aneurysm (AAA). Tissue inhibitor of metalloproteinases-4 (TIMP-4) expression is reduced in AAA patients. In this study, we investigated the impact of TIMP-4 on the phenotype alterations induced by angiotensin II (Ang-II) in human vascular smooth muscle cells (VSMCs). Methods: The expression profiling of TIMP-4 and A-kinase anchoring protein (AKAP1) in AAA samples was analyzed using the GSE7084 and GSE140947 datasets. Levels of TIMP-4 and AKAP1 in Ang-II-exposed VSMCs and AAA tissues and serum samples were detected. RNA immunoprecipitation (RIP) experiment and mRNA stability analysis were used to examine the interaction between AKAP1 and TIMP-4 mRNA. The impact of the AKAP1/TIMP-4 cascade on Ang-II-induced VSMC phenotype alterations was determined by evaluating cell viability, apoptosis, oxidative stress, and inflammation. Results: TIMP-4 and AKAP1 levels were decreased in Ang-II-exposed VSMCs. Increased TIMP-4 expression protected VSMCs against Ang-II-evoked growth impairment in vitro . Moreover, TIMP-4 upregulation diminished Ang-II-evoked oxidative stress and inflammation in VSMCs. Mechanistically, RNA binding protein (RBP) AKAP1 stabilized TIMP-4 mRNA to elevate TIMP-4 expression. TIMP-4 reduction partially abrogated AKAP1-driven suppression on oxidative stress, inflammation, matrix metalloproteinase (MMP9) expression, and nuclear factor kappa B (NF-κB) pathway activation in Ang-II-exposed VSMCs. Additionally, TIMP-4 and AKAP1 levels were downregulated in AAA patients in their AAA tissues and serum samples. TIMP-4 and AKAP1 had good diagnostic values for AAA with high area under the ROC curve. Conclusion: Our study provides evidence for the role of the AKAP1/TIMP-4/NF-κB pathway in Ang-II-induced VSMC inflammation and oxidative stress.
Collapse
Affiliation(s)
- Yongxin Han
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital, Jinan City, Shandong, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Javed MJ, Howard RM, Li H, Carrasco L, Dirain ML, Su G, Cai G, Upchurch GR, Jiang Z. GSDMD Deficiency Attenuates the Development of Ascending Aortic Dissections in a Novel Mouse Model. Arterioscler Thromb Vasc Biol 2025; 45:541-556. [PMID: 39945067 PMCID: PMC11945581 DOI: 10.1161/atvbaha.124.321740] [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: 08/21/2024] [Accepted: 01/27/2025] [Indexed: 02/19/2025]
Abstract
BACKGROUND Mechanisms driving the development of type A aortic dissection (TAD) are currently poorly understood, and animal models of spontaneous TAD are limited. In the present study, we developed a novel mouse TAD model and evaluated the role of GSDMD (gasdermin D) in TAD development. METHODS TADs were created by treating the ascending aorta of adult C57BL/6J mice with Act E (active elastase) and β-aminopropionitrile. The temporal progress of the TAD pathology was rigorously characterized by histological evaluation and scanning electron microscopy, while potential mechanisms were explored using bulk RNA sequencing of specimens collected at multiple time points. With this novel TAD model, we conducted additional experiments to investigate the impact of GSDMD deficiency on TAD formation. RESULTS Ascending aortas challenged with Act E and β-aminopropionitrile developed pathology featuring the early onset of intimomedial tears (complete penetration) and intramural hematomas, followed by progressive medial loss and aortic dilation. Ingenuity pathway analysis and functional annotation of differentially expressed genes suggested that a unique inflammatory microenvironment, rather than general inflammation, promotes the onset of TADs by specifically recruiting neutrophils to the aortic wall. At later stages, T cell-mediated immune injury emerged as the primary driver of pathology. Gsdmd deficiency attenuated medial loss, adventitial fibrosis, and dilation of TADs. This protective effect correlated with a reduced cell death and decreased T-cell infiltration in TADs. Notably, cleaved GSDMD was detected in human TADs but was absent in healthy aortas. CONCLUSIONS A novel mouse TAD model was developed, specifically targeting the ascending aorta. This model generates a unique microenvironment that activates specific immune cell subsets, driving the onset and subsequent remodeling of TADs. Consistently, Gsdmd deficiency mitigates TAD development, likely by modulating cell death and T-cell responses. This model provides a valuable tool for studying immune injury mechanisms in TAD pathogenesis.
Collapse
Affiliation(s)
- Muhammad J. Javed
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Rachael M. Howard
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Hua Li
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Laura Carrasco
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Marvin L.S Dirain
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Gang Su
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Guoshuai Cai
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Gilbert R. Upchurch
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Zhihua Jiang
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
- Malcom Randll VA Medical Center, Gainesville, FL 32608
| |
Collapse
|
3
|
Dai F, Cao Y, Zhu C, Li Y, Ma X, Wang S, Liu H, Xie X, Gao L, Wang Y, Wang C. Design, Synthesis, and Biological Evaluation of Naphthalimide-Polyamine Conjugate as a Potential Anti-Colorectal Cancer Agent. Chem Biodivers 2025; 22:e202401873. [PMID: 39632400 DOI: 10.1002/cbdv.202401873] [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: 08/03/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
Colorectal cancer is the second most common cause of cancer-related death worldwide, with a rising incidence, highlighting an urgent need for novel therapeutics. In this study, we developed several polyamine conjugates. Compound 6 (C6) was selected as the lead compound and was evaluated for anticancer activity in vitro and in vivo. In vitro experiments showed that C6 inhibited cell proliferation, colony formation, migration, and invasion of colorectal cancer cells while inducing apoptosis, pyroptosis, and autophagosome formation. Genetic or pharmacological inhibition of autophagy weakened C6-induced apoptosis and gasdermin E (GSDME)-dependent pyroptosis. Inactivation of caspase 3 activity by AC-DEVD-CHO decreased the levels of N-terminal GSDME induced by C6. Furthermore, animal models exhibited suppressed tumor growth and dissemination after treatment with C6. Taken together, our findings highlight C6 as a potential drug against colorectal cancer.
Collapse
Affiliation(s)
- Fujun Dai
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Yue Cao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Chenguang Zhu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Yibing Li
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Xiaoxuan Ma
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Senzhen Wang
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Haizhen Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Xiaoya Xie
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Lei Gao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Yanming Wang
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Chaojie Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| |
Collapse
|
4
|
Cai H, Li H, Xiao X, Wang S, Liu R, Qin Y, Zhou Y, Yao C. TRAF6 promotes abdominal aortic aneurysm development by activating macrophage pyroptosis via the NLRP3/Caspase1/GSDMD pathway. FASEB J 2025; 39:e70318. [PMID: 39831511 DOI: 10.1096/fj.202402873r] [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: 11/17/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Abdominal aortic aneurysm represents a critical pathology of the aorta that currently lacks effective pharmacological interventions. TNF receptor-associated factor 6 (TRAF6) has been established to be involved in cardiovascular diseases such as atherosclerosis, hypertension, and heart failure. However, its role in abdominal aortic aneurysm (AAA) remains unclear. This study aimed to explore the role of TRAF6 on AAA formation and its underlying mechanisms. Single-cell RNA sequencing of human AAA tissues demonstrated that TRAF6 was significantly upregulated in aortic macrophages. Moreover, overexpression of TRAF6 promotes AAA formation in elastase-induced C57BL/6 mice, while TRAF6 pharmacological inhibition could attenuate AAA development. Consistently, inhibition of TRAF6 in macrophages through in vitro methods notably limits their pyroptosis, while also diminishing proinflammatory responses in these cells. Mechanistically, TRAF6 can modulate macrophage pyroptosis through the NLRP3/Caspase1/GSDMD signaling pathway. Our study highlights the crucial role of the TRAF6/NLRP3/Caspase1/GSDMD axis in macrophage pyroptosis and AAA, offering potential biomarkers and therapeutic targets for AAA.
Collapse
Affiliation(s)
- Huoying Cai
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Huaming Li
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaoyong Xiao
- Department of Emergency Medicine, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Siwen Wang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Ruiming Liu
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Yuansen Qin
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Yu Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Chen Yao
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Jia W, Chan JC, Wong TY, Fisher EB. Diabetes in China: epidemiology, pathophysiology and multi-omics. Nat Metab 2025; 7:16-34. [PMID: 39809974 DOI: 10.1038/s42255-024-01190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Although diabetes is now a global epidemic, China has the highest number of affected people, presenting profound public health and socioeconomic challenges. In China, rapid ecological and lifestyle shifts have dramatically altered diabetes epidemiology and risk factors. In this Review, we summarize the epidemiological trends and the impact of traditional and emerging risk factors on Chinese diabetes prevalence. We also explore recent genetic, metagenomic and metabolomic studies of diabetes in Chinese, highlighting their role in pathogenesis and clinical management. Although heterogeneity across these multidimensional areas poses major analytic challenges in classifying patterns or features, they have also provided an opportunity to increase the accuracy and specificity of diagnosis for personalized treatment and prevention. National strategies and ongoing research are essential for improving diabetes detection, prevention and control, and for personalizing care to alleviate societal impacts and maintain quality of life.
Collapse
Affiliation(s)
- Weiping Jia
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute for Proactive Healthcare, Shanghai Jiao Tong University, Shanghai, China.
| | - Juliana Cn Chan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences and Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Tien Y Wong
- Tsinghua Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Singapore National Eye Center, SingHealth, Singapore, Singapore
| | - Edwin B Fisher
- Peers for Progress, Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Liu SJ, Yang XQ, Lu HQ, Zhang KC, Tang YJ, Xu Y. A bibliometric analysis of abdominal aortic aneurysm (2014-2024). Front Cardiovasc Med 2024; 11:1436600. [PMID: 39735864 PMCID: PMC11672341 DOI: 10.3389/fcvm.2024.1436600] [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: 05/22/2024] [Accepted: 11/29/2024] [Indexed: 12/31/2024] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a localized bulge of the abdominal aorta, which mainly manifests as a pulsatile mass in the abdomen. Once an abdominal aortic aneurysm ruptures, the patient's life is seriously endangered. Surgery is the preferred treatment for abdominal aortic aneurysm. At present, there has been no comprehensive review of the current status of abdominal aortic aneurysm research. Therefore, this study aimed to identify global trends in abdominal aortic aneurysm research over the last 10 years through bibliometric analysis and to inform clinical practice, research funding allocation, and decision-making. Methods We downloaded research articles and reviews on abdominal aortic aneurysm from 1 January 2014, to 1 March 2024, from the Web of Science core collection. CiteSpace (version 6.2.1), RStudio and VOSviewer (version 1.6.18) were used for visual analysis of regional distribution, institutions, authors, keywords and other information. Results The number of documents on abdominal aortic aneurysm research increased continuously and has stabilized in recent years. A total of 9,905 publications from 67 countries were published from 1 January 2014, to 1 March 2024. A total of 2,142 (29.52%) studies were from the United States, 1,293 (13.05%) were from China, and 919 (9.28%) were from the United Kingdom. A total of 205 studies were conducted at Stanford University, 172 were conducted at Harvard Medical School, and 165 were conducted at the Mayo Clinic. The top three coauthorship authors were Schermerhorn, Marc L (114); Golledge, Jonathan (102); and De Vries, Jean Paul P.M. (74). The most cocited reference was Chaikof EL, 2018, J Vasc Surg, v67, p. 2; the most cocited journal was the Journal of Vascular Surgery; and the most cocited author was Lederle, FA. "Abdominal aortic aneurysm" was the most frequently used author keyword (2,492). Twenty-five references with strong citation bursts were identified by "CiteSpace". "Artificial intelligence", "clinical outcomes" and "bridging stent" were the primary keywords of emerging research hotspots. Conclusion This is the first bibliometric study to comprehensively summarize the research trends in abdominal aortic aneurysm research. This information can help us to identify the current research hotspots and directions. This study will provide extensive help for future research.
Collapse
Affiliation(s)
- Shao-Jia Liu
- Panzhihua Central Hospital, Panzhihua, Sichuan, China
| | - Xin-Qing Yang
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Hong-Qiao Lu
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Kun-Chi Zhang
- Panzhihua Central Hospital, Panzhihua, Sichuan, China
| | | | - Yu Xu
- Panzhihua Central Hospital, Panzhihua, Sichuan, China
| |
Collapse
|
7
|
Hu Y, Feng Z, An G, Lv Z, Wang J, Cui Y, Corrigan CJ, Wang W, Li Q, Ying S. Edwardsiella tarda induces airways inflammation and production of autoantibodies against lung tissues through regulation of the IL-33-ST2 axis. Immunology 2024; 173:575-589. [PMID: 39126327 DOI: 10.1111/imm.13848] [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: 12/16/2023] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent chronic respiratory disease characterised by irreversible airways obstruction associated with chronic airways inflammation and remodelling, while the pathogenesis and the mechanistic differences between patients remain to be fully elucidated. We previously reported that alarmin cytokine IL-33 may contribute to the production of autoantibodies against respiratory epithelial cells. Here we expand the hypothesis that pulmonary autoimmune responses induced by airway microbiota also contribute to the progression of COPD. We focused on Edwardsiella tarda which we detected uniquely in the induced sputum of patients with acute exacerbations of COPD. Pernasal challenge of the airways of WT mice with supernatants of cultured E. tarda induced marked, elevated expression of IL-33 in the lung tissues. Immunisation of animals with supernatants of cultured E. tarda resulted in significantly elevated airways inflammation, the formation of tertiary lymphatic structures and significantly elevated proportions of T follicular helper T cells in the lung tissue and mediastinal lymph nodes. Interestingly, such challenge also induced production of IgG autoantibodies directed against lung tissue lysate, alveolar epithelial cell proteins and elastin fragment, while putrescine, one of metabolites generated by the bacterium, might play an important role in the autoantibody production. Furthermore, all of these effects were partly but significantly abrogated in mice with deletion of the IL-33 receptor ST2. Collectively, these data support the hypothesis that COPD is progressed at least partly by airways microbiota such as E. tarda initiating autoimmune attack of the airways epithelium mediated at least partly through the IL-33-ST2 axis.
Collapse
Affiliation(s)
- Yue Hu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhihong Feng
- Department of Respiratory Medicine, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Gao An
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Wang
- Department of Laboratory Animal Sciences, Capital Medical University, Beijing, China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chris J Corrigan
- Faculty of Life Sciences & Medicine, School of Immunology & Microbial Sciences, Department of Inflammation Biology, King's College London, London, UK
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qin Li
- Department of Laboratory, Yanjing Medical College, Capital Medical University, Beijing, China
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Li Z, Chen L, Qu L, Yu W, Liu T, Ning F, Li J, Guo X, Sun F, Sun B, Luo L. Potential implications of natural compounds on aging and metabolic regulation. Ageing Res Rev 2024; 101:102475. [PMID: 39222665 DOI: 10.1016/j.arr.2024.102475] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Aging is generally accompanied by a progressive loss of metabolic homeostasis. Targeting metabolic processes is an attractive strategy for healthy-aging. Numerous natural compounds have demonstrated strong anti-aging effects. This review summarizes recent findings on metabolic pathways involved in aging and explores the anti-aging effects of natural compounds by modulating these pathways. The potential anti-aging effects of natural extracts rich in biologically active compounds are also discussed. Regulating the metabolism of carbohydrates, proteins, lipids, and nicotinamide adenine dinucleotide is an important strategy for delaying aging. Furthermore, phenolic compounds, terpenoids, alkaloids, and nucleotide compounds have shown particularly promising effects on aging, especially with respect to metabolism regulation. Moreover, metabolomics is a valuable tool for uncovering potential targets against aging. Future research should focus on identifying novel natural compounds that regulate human metabolism and should delve deeper into the mechanisms of metabolic regulation using metabolomics methods, aiming to delay aging and extend lifespan.
Collapse
Affiliation(s)
- Zhuozhen Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Lili Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; School of Life Science, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Liangliang Qu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Wenjie Yu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Tao Liu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Fangjian Ning
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jinwang Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Xiali Guo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Fengjie Sun
- Department of Biological Sciences, School of Science and Technology, Georgia Gwinnett College, Lawrenceville, GA 30043, USA
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Liping Luo
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
9
|
Yin Z, Zhang J, Shen Z, Qin J, Wan J, Wang M. Regulated vascular smooth muscle cell death in vascular diseases. Cell Prolif 2024; 57:e13688. [PMID: 38873710 PMCID: PMC11533065 DOI: 10.1111/cpr.13688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024] Open
Abstract
Regulated cell death (RCD) is a complex process that involves several cell types and plays a crucial role in vascular diseases. Vascular smooth muscle cells (VSMCs) are the predominant elements of the medial layer of blood vessels, and their regulated death contributes to the pathogenesis of vascular diseases. The types of regulated VSMC death include apoptosis, necroptosis, pyroptosis, ferroptosis, parthanatos, and autophagy-dependent cell death (ADCD). In this review, we summarize the current evidence of regulated VSMC death pathways in major vascular diseases, such as atherosclerosis, vascular calcification, aortic aneurysm and dissection, hypertension, pulmonary arterial hypertension, neointimal hyperplasia, and inherited vascular diseases. All forms of RCD constitute a single, coordinated cell death system in which one pathway can compensate for another during disease progression. Pharmacologically targeting RCD pathways has potential for slowing and reversing disease progression, but challenges remain. A better understanding of the role of regulated VSMC death in vascular diseases and the underlying mechanisms may lead to novel pharmacological developments and help clinicians address the residual cardiovascular risk in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Zican Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Juan‐Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Center for Healthy Aging, Wuhan University School of NursingWuhanChina
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
10
|
Su Y, Lu C, Chen S. Construction of the miRNA/Pyroptosis-Related Molecular Regulatory Axis in Abdominal Aortic Aneurysm: Evidence From Transcriptome Data Combined With Multiple Machine Learning Approaches Followed by Experiment Validation. J Immunol Res 2024; 2024:1429510. [PMID: 39512836 PMCID: PMC11540895 DOI: 10.1155/2024/1429510] [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: 06/05/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Background: Abdominal aortic aneurysm (AAA) represents a permanent and localized widening of the abdominal aorta, posing a potentially lethal risk of aortic rupture. Several recent studies have highlighted the role of pyroptosis, a pro-inflammatory programed cell death, as critical molecular regulators in AAA occurrence, progression, and rupture. However, the potential effects of pyroptosis in AAA and its upstream microRNA (miRNA) have not been comprehensively clarified. Methods: Through a search of the gene expression omnibus (GEO) database, the expression profiles of mRNAs (GSE7084, GSE57691, and GSE98278) and miRNAs (GSE62179) and corresponding clinical features were downloaded, respectively. Expression profiles of 15 AAA and 10 normal vascular samples were consecutively collected for in vitro experimentation and subsequent analysis. Various machine learning techniques were employed to identify hub pyroptosis-related genes (PRGs), leading to the development of a predictive model termed the PRG classifier. Quantitative real-time-polymerase chain reaction (qRT-PCR), western blot (WB), and enzyme-linked immunosorbent assay (ELISA) were used to confirm the expression of the hub PRGs. The diagnostic and predictive capabilities of the model were comprehensively evaluated in GEO and hospital cohorts. Then, the crucial immune cell infiltration and molecular pathways implicated in the initiation and rupture of AAA and their association with pyroptosis were explored. Lastly, a miRNA/hub pyroptosis-related molecular regulatory axis was constructed using the TargetScan dataset, which was further explored through loss-of-function assays. Results: Differential analysis, enrichment score analysis, and principal component analysis (PCA) revealed that pyroptosis-related molecules were significantly involved in the occurrence of AAA. Utilizing multiple machine learning algorithms, eight key PRGs (cysteinyl aspartate specific proteinase [CASP]1, infiltrating lymphocyte [IL]1B, IL18, IL6, NOD-, LRR- and pyrin domain-containing protein [NLRP]1, NLRP2, NLRP3, and tumor necrosis factor [TNF]) were integrated to establish a PRG classifier. Demonstrating robust diagnostic capabilities (area under curve [AUC] > 0.90), the PRG classifier provided clinical insights across two GEO datasets and effectively differentiated small AAA from large AAA, elective stable AAA (eAAA), and ruptured AAA (rAAA), respectively. qRT-PCR, WB, and ELISA verified the mRNA and protein expression of the hub PRGs. Notably, in hospital cohorts, a substantial positive link was unveiled between the PRG classifier and AAA risk factors (hypertension history, diastolic pressure, triglyceride levels, and aneurysm diameter). Furthermore, immune cell infiltration and functional enrichment analysis revealed significant associations of the PRG classifier/PRGs with M2 macrophage infiltration, activated dendritic cells, and enrichment scores of the cytosolic deoxyribonucleic acid (DNA) sensing pathway and tryptophan metabolism, potentially mediating AAA onset and rupture. Finally, based on 90 differentially expressed miRNAs (DEmiRNAs) and eight hub PRGs through TargetScan dataset, a hsa-miR-331-3p/TNF regulatory axis was constructed, wherein upregulation of hsa-miR-331-3p expression significantly reduced TNF and CASP1 protein levels. Conclusion: A predictive model (PRG classifier) incorporating eight PRGs through multiple machine learning algorithms was developed and validated. This model may stand as a potent tool for diagnosing AAA and assessing disease severity. The identification of the cytosolic DNA sensing pathway and the hsa-miR-331-3p/TNF interaction axis may represent crucial targets for AAA treatment, offering deeper insights into its potential pathogenesis.
Collapse
Affiliation(s)
- Yongchao Su
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Chuangang Lu
- Department of Cardiothoracic Surgery, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya 572000, Hainan Province, China
| | - Shuchen Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
11
|
Wang J, Ye W, Zou J, Yang P, Jin M, Zheng Z, Zhou C, Qiu W, Lu J, Li C, Guo S, Xu Y, Huang Z, Liu P, Liu Z. Targeting the smooth muscle cell Keap1-Nrf2-GSDMD-pyroptosis axis by cryptotanshinone prevents abdominal aortic aneurysm formation. Theranostics 2024; 14:6516-6542. [PMID: 39479449 PMCID: PMC11519792 DOI: 10.7150/thno.98400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024] Open
Abstract
Rationale: Abdominal aortic aneurysm (AAA) is an inflammatory, fatal aortic disease that currently lacks any effective drugs. Cryptotanshinone (CTS) is a prominent and inexpensive bioactive substance derived from Salvia miltiorrhiza Bunge, a well-known medicinal herb for treating cardiovascular diseases through its potent anti-inflammatory properties. Nevertheless, the therapeutic effect of CTS on AAA formation remains unknown. Methods: To investigate the therapeutic effect of CTS in AAA, variety of experimental approaches were employed, majorly including AAA mouse model establishment, real-time polymerase chain reaction (PCR), RNA sequencing, western blot, co-immunoprecipitation, scanning/transmission electron microscopy (SEM/TEM), enzyme-linked immunosorbent assay (ELISA), seahorse analysis, immunohistochemistry, and confocal imaging. Results: In this study, we demonstrated that CTS suppressed the formation of AAA in apolipoprotein E knock-out (ApoE-/-) mice infused with Ang II. A combination of network pharmacology and whole transcriptome sequencing analysis indicated that activation of the Keap1-Nrf2 pathway and regulation of programmed cell death in vascular smooth muscle cells (VSMCs) are closely linked to the anti-AAA effect of CTS. Mechanistically, CTS promoted the transcription of Nrf2 target genes, particularly Hmox-1, which prevented the activation of NLRP3 and GSDMD-initiated pyroptosis in VSMCs, thereby mitigating VSMC inflammation and maintaining the VSMC contractile phenotype. Subsequently, by utilizing molecular docking, together with the cellular thermal shift assay (CETSA) and isothermal titration calorimetry (ITC), a particular binding site was established between CTS and Keap1 at Arg415. To confirm the binding site, site-directed mutagenesis was performed, which intriguingly showed that the Arg415 mutation eliminated the binding between CTS and the Keap1-Nrf2 protein and abrogated the antioxidant and anti-pyroptosis effects of CTS. Furthermore, VSMC-specific Nrf2 knockdown in mice dramatically reversed the protective action of CTS in AAA and the inhibitory effect of CTS on VSMC pyroptosis. Conclusion: Naturally derived CTS exhibits promising efficacy as a treatment drug for AAA through its targeting of the Keap1-Nrf2-GSDMD-pyroptosis axis in VSMCs.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/pathology
- Disease Models, Animal
- Kelch-Like ECH-Associated Protein 1/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NF-E2-Related Factor 2/metabolism
- Phenanthrenes/pharmacology
- Pyroptosis/drug effects
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Chunhong Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Wanlu Qiu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chengzhi Li
- Department of Interventional Radiology and Vascular Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Shuai Guo
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yiming Xu
- School of Basic Medical Sciences, State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Peiqing Liu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511443, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 511443, China
| |
Collapse
|
12
|
Cui Y, Tan C, Zhang W, Jiang P, Sun J, Mei F. Establishment of Mouse Models of Abdominal Aortic Aneurysm. Angiology 2024:33197241284848. [PMID: 39268808 DOI: 10.1177/00033197241284848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular disease that commonly affects elderly individuals but has recently increased in younger populations. As the aneurysm grows, it can cause compression symptoms such as abdominal pain, rupture, and bleeding, which are absent in the early stages. Once an AAA ruptures and causes bleeding, the mortality rate is alarmingly high. Currently, the pathogenesis for AAA is unknown, and therapeutic options are limited, necessitating improvement in treatment efficacy. An essential research method for studying the processes and potential treatment of AAA is establishing animal models using mice. The present study provides a detailed overview of the widely used AAA mouse animal models and their construction strategies, advantages, disadvantages, scope of applications, and prospects.
Collapse
Affiliation(s)
- Yongpan Cui
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Chengpeng Tan
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Wuming Zhang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Peng Jiang
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Jianfeng Sun
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| | - Fei Mei
- Department of Vascular Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Hubei, China
| |
Collapse
|
13
|
Javed MJ, Howard RM, Li H, Carrasco L, Dirain MLS, Su G, Cai G, Upchurch GR, Jiang Z. Gasdermin D deficiency attenuates development of ascending aortic dissections in a novel mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609270. [PMID: 39229014 PMCID: PMC11370574 DOI: 10.1101/2024.08.22.609270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Thoracic aortic dissection (TAD) is a silent killer. Approximately two-thirds of the cases occur in the ascending aorta (i.e. type A dissection) and majority of them are unrelated to genetic mutations. However, animal models of spontaneous type A dissection are not widely available. In the present study, a novel mouse TAD model was created. Further, the role of gasdermin D (GSDMD) in TAD development was evaluated. Methods TADs were created by treating ascending aorta of adult mice (C57BL/6J) with active elastase (40.0 U/ml) and β-aminopropionitrile (Act E+BAPN). The temporal progress of the TAD pathology was rigorously characterized by histological evaluation and scanning electron microscopy, while potential mechanisms explored with bulk RNA sequencing of specimens collected at multiple timepoints. With this novel TAD model, further experiments were performed with Gsdmd -/- mice to evaluate its impact on TAD formation. Results The ascending aorta challenged with Act E+BAPN developed pathology characterized by an early onset of intimomedial tears (complete penetration) and intramural hematoma, followed by progressive medial loss and aortic dilation. Ingenuity Pathway Analysis and functional annotation of differentially expressed genes suggested that a unique inflammatory micro-environment, rather than general inflammation, promoted the onset of TADs by specifically recruiting neutrophils to the aortic wall, while the pathology at the advanced stage was driven by T-cell mediated immune injury. Gsdmd -/- attenuated medial loss, adventitial fibrosis, and dilation of TADs. This protective effect was associated with a reduced number of TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) positive cells and T-cells in TADs. Conclusions A novel mouse TAD model was created in the ascending aorta. It produces a unique microenvironment to activate different immune cell subsets, promoting onset and subsequent remodeling of TADs. Consistently, Gsdmd -/- attenuates TAD development, with modulation of cell death and T-cell response likely acting as the underlying mechanism.
Collapse
|
14
|
Wu Z, Xu Z, Pu H, Ding A, Hu J, Lei J, Zeng C, Qiu P, Qin J, Wu X, Li B, Wang X, Lu X. NINJ1 Facilitates Abdominal Aortic Aneurysm Formation via Blocking TLR4-ANXA2 Interaction and Enhancing Macrophage Infiltration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306237. [PMID: 38922800 PMCID: PMC11336960 DOI: 10.1002/advs.202306237] [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: 08/31/2023] [Revised: 01/30/2024] [Indexed: 06/28/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a common and potentially life-threatening condition. Chronic aortic inflammation is closely associated with the pathogenesis of AAA. Nerve injury-induced protein 1 (NINJ1) is increasingly acknowledged as a significant regulator of the inflammatory process. However, the precise involvement of NINJ1 in AAA formation remains largely unexplored. The present study finds that the expression level of NINJ1 is elevated, along with the specific expression level in macrophages within human and angiotensin II (Ang II)-induced murine AAA lesions. Furthermore, Ninj1flox/flox and Ninj1flox/floxLyz2-Cre mice on an ApoE-/- background are generated, and macrophage NINJ1 deficiency inhibits AAA formation and reduces macrophage infiltration in mice infused with Ang II. Consistently, in vitro suppressing the expression level of NINJ1 in macrophages significantly restricts macrophage adhesion and migration, while attenuating macrophage pro-inflammatory responses. Bulk RNA-sequencing and pathway analysis uncover that NINJ1 can modulate macrophage infiltration through the TLR4/NF-κB/CCR2 signaling pathway. Protein-protein interaction analysis indicates that NINJ1 can activate TLR4 by competitively binding with ANXA2, an inhibitory interacting protein of TLR4. These findings reveal that NINJ1 can modulate AAA formation by promoting macrophage infiltration and pro-inflammatory responses, highlighting the potential of NINJ1 as a therapeutic target for AAA.
Collapse
Affiliation(s)
- Zhaoyu Wu
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
- Vascular Center of Shanghai JiaoTong UniversityShanghai200011China
| | - Zhijue Xu
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
- Key Laboratory of Systems Biomedicine (Ministry of Education)Shanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Hongji Pu
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Ang'ang Ding
- Department of UltrasoundShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Jiateng Hu
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Jiahao Lei
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Chenlin Zeng
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Peng Qiu
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
- Vascular Center of Shanghai JiaoTong UniversityShanghai200011China
| | - Jinbao Qin
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
- Vascular Center of Shanghai JiaoTong UniversityShanghai200011China
| | - Xiaoyu Wu
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
- Vascular Center of Shanghai JiaoTong UniversityShanghai200011China
| | - Bo Li
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
| | - Xin Wang
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
- Vascular Center of Shanghai JiaoTong UniversityShanghai200011China
| | - Xinwu Lu
- Department of Vascular SurgeryShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011China
- Vascular Center of Shanghai JiaoTong UniversityShanghai200011China
| |
Collapse
|
15
|
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
|
16
|
Zhu C, Xu S, Jiang R, Yu Y, Bian J, Zou Z. The gasdermin family: emerging therapeutic targets in diseases. Signal Transduct Target Ther 2024; 9:87. [PMID: 38584157 PMCID: PMC10999458 DOI: 10.1038/s41392-024-01801-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
The gasdermin (GSDM) family has garnered significant attention for its pivotal role in immunity and disease as a key player in pyroptosis. This recently characterized class of pore-forming effector proteins is pivotal in orchestrating processes such as membrane permeabilization, pyroptosis, and the follow-up inflammatory response, which are crucial self-defense mechanisms against irritants and infections. GSDMs have been implicated in a range of diseases including, but not limited to, sepsis, viral infections, and cancer, either through involvement in pyroptosis or independently of this process. The regulation of GSDM-mediated pyroptosis is gaining recognition as a promising therapeutic strategy for the treatment of various diseases. Current strategies for inhibiting GSDMD primarily involve binding to GSDMD, blocking GSDMD cleavage or inhibiting GSDMD-N-terminal (NT) oligomerization, albeit with some off-target effects. In this review, we delve into the cutting-edge understanding of the interplay between GSDMs and pyroptosis, elucidate the activation mechanisms of GSDMs, explore their associations with a range of diseases, and discuss recent advancements and potential strategies for developing GSDMD inhibitors.
Collapse
Affiliation(s)
- Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ruoyu Jiang
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
17
|
Yang Y, Zhang H, Wang Y, Xu J, Shu S, Wang P, Ding S, Huang Y, Zheng L, Yang Y, Xiong C. Promising dawn in the management of pulmonary hypertension: The mystery veil of gut microbiota. IMETA 2024; 3:e159. [PMID: 38882495 PMCID: PMC11170974 DOI: 10.1002/imt2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/15/2023] [Accepted: 11/25/2023] [Indexed: 06/18/2024]
Abstract
The gut microbiota is a complex community of microorganisms inhabiting the intestinal tract, which plays a vital role in human health. It is intricately involved in the metabolism, and it also affects diverse physiological processes. The gut-lung axis is a bidirectional pathway between the gastrointestinal tract and the lungs. Recent research has shown that the gut microbiome plays a crucial role in immune response regulation in the lungs and the development of lung diseases. In this review, we present the interrelated factors concerning gut microbiota and the associated metabolites in pulmonary hypertension (PH), a lethal disease characterized by elevated pulmonary vascular pressure and resistance. Our research team explored the role of gut-microbiota-derived metabolites in cardiovascular diseases and established the correlation between metabolites such as putrescine, succinate, trimethylamine N-oxide (TMAO), and N, N, N-trimethyl-5-aminovaleric acid with the diseases. Furthermore, we found that specific metabolites, such as TMAO and betaine, have significant clinical value in PH, suggesting their potential as biomarkers in disease management. In detailing the interplay between the gut microbiota, their metabolites, and PH, we underscored the potential therapeutic approaches modulating this microbiota. Ultimately, we endeavor to alleviate the substantial socioeconomic burden associated with this disease. This review presents a unique exploratory analysis of the link between gut microbiota and PH, intending to propel further investigations in the gut-lung axis.
Collapse
Affiliation(s)
- Yicheng Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Hanwen Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yaoyao Wang
- State Key Laboratory of Cardiovascular Disease, Department of Nephrology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Department of Genetics University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Songren Shu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Peizhi Wang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Center for Molecular Cardiology University of Zurich Zurich Switzerland
| | - Shusi Ding
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
| | - Yuan Huang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Lemin Zheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
- Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, School of Basic Medical Sciences, Health Science Center The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, Peking University Beijing China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Changming Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
18
|
Dai Y, Wei X, Jiang T, Wang Q, Li Y, Ruan N, Luo P, Huang J, Yang Y, Yan Q, Zhang C, Liu Y. Ferroptosis in age-related vascular diseases: Molecular mechanisms and innovative therapeutic strategies. Biomed Pharmacother 2024; 173:116356. [PMID: 38428313 DOI: 10.1016/j.biopha.2024.116356] [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: 01/08/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
Aging, an inevitable aspect of human existence, serves as one of the predominant risk factors for vascular diseases. Delving into the mystery of vascular disease's pathophysiology, the profound involvement of programmed cell death (PCD) has been extensively demonstrated. PCD is a fundamental biological process that plays a crucial role in both normal physiology and pathology, including a recently discovered form, ferroptosis. Ferroptosis is characterized by its reliance on iron and lipid peroxidation, and its significant involvement in vascular disease pathophysiology has been increasingly acknowledged. This phenomenon not only offers a promising therapeutic target but also deepens our understanding of the complex relationship between ferroptosis and age-related vascular diseases. Consequently, this article aims to thoroughly review the mechanisms that enable the effective control and inhibition of ferroptosis. It focuses on genetic and pharmacological interventions, with the goal of developing innovative therapeutic strategies to combat age-related vascular diseases.
Collapse
Affiliation(s)
- Yue Dai
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiuxian Wei
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Wang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Li
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Nan Ruan
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pengcheng Luo
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingwen Huang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Nursing, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Yang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Yan
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Liu
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
19
|
Hu RT, Deng HW, Teng WB, Zhou SD, Ye ZM, Dong ZM, Qin C. ADORA3: A Key Player in the Pathogenesis of Intracranial Aneurysms and a Potential Diagnostic Biomarker. Mol Diagn Ther 2024; 28:225-235. [PMID: 38341835 DOI: 10.1007/s40291-024-00694-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND The effects of genes on the development of intracranial aneurysms (IAs) remain to be elucidated, and reliable blood biomarkers for diagnosing IAs are yet to be established. This study aimed to identify genes associated with IAs pathogenesis and explore their diagnostic value by analyzing IAs datasets, conducting vascular smooth muscle cells (VSMC) experiments, and performing blood detection. METHODS IAs datasets were collected and the differentially expressed genes were analyzed. The selected genes were validated in external datasets. Autophagy was induced in VSMC and the effect of selected genes was determined. The diagnostic value of selected gene on the IAs were explored using area under curve (AUC) analysis using IAs plasma samples. RESULTS Analysis of 61 samples (32 controls and 29 IAs tissues) revealed a significant increase in expression of ADORA3 compared with normal tissues using empirical Bayes methods of "limma" package; this was further validated by two external datasets. Additionally, induction of autophagy in VSMC lead to upregulation of ADORA3. Conversely, silencing ADORA3 suppressed VSMC proliferation and autophagy. Furthermore, analysis of an IAs blood sample dataset and clinical plasma samples demonstrated increased ADORA3 expression in patients with IA compared with normal subjects. The diagnostic value of blood ADORA3 expression in IAs was moderate when analyzing clinical samples (AUC: 0.756). Combining ADORA3 with IL2RB or CCR7 further enhanced the diagnostic ability for IAs, with the AUC value over 0.83. CONCLUSIONS High expression of ADORA3 is associated with IAs pathogenesis, likely through its promotion of VSMC autophagy. Furthermore, blood ADORA3 levels have the potential to serve as an auxiliary diagnostic biomarker for IAs.
Collapse
Affiliation(s)
- Rui-Ting Hu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Hao-Wei Deng
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Wen-Bin Teng
- Department of Neurology, Minzu Hospital of Guangxi Medical University, Nanning, 530001, China
| | - Shao-Dan Zhou
- Department of Neurology, Minzu Hospital of Guangxi Medical University, Nanning, 530001, China
| | - Zi-Ming Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Zi-Mei Dong
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Chao Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China.
| |
Collapse
|
20
|
Yin Z, Zhang J, Zhao M, Peng S, Ye J, Liu J, Xu Y, Xu S, Pan W, Wei C, Qin J, Wan J, Wang M. Maresin-1 ameliorates hypertensive vascular remodeling through its receptor LGR6. MedComm (Beijing) 2024; 5:e491. [PMID: 38463394 PMCID: PMC10924638 DOI: 10.1002/mco2.491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/30/2023] [Accepted: 01/11/2024] [Indexed: 03/12/2024] Open
Abstract
Hypertensive vascular remodeling is defined as the changes in vascular function and structure induced by persistent hypertension. Maresin-1 (MaR1), one of metabolites from Omega-3 fatty acids, has been reported to promote inflammation resolution in several inflammatory diseases. This study aims to investigate the effect of MaR1 on hypertensive vascular remodeling. Here, we found serum MaR1 levels were reduced in hypertensive patients and was negatively correlated with systolic blood pressure (SBP). The treatment of MaR1 reduced the elevation of blood pressure and alleviated vascular remodeling in the angiotensin II (AngII)-infused mouse model. In addition, MaR1-treated vascular smooth muscle cells (VSMCs) exhibited reduced excessive proliferation, migration, and phenotype switching, as well as impaired pyroptosis. However, the knockout of the receptor of MaR1, leucine-rich repeat-containing G protein-coupled receptor 6 (LGR6), was seen to aggravate pathological vascular remodeling, which could not be reversed by additional MaR1 treatment. The mechanisms by which MaR1 regulates vascular remodeling through LGR6 involves the Ca2+/calmodulin-dependent protein kinase II/nuclear factor erythroid 2-related factor 2/heme oxygenase-1 signaling pathway. Overall, supplementing MaR1 may be a novel therapeutic strategy for the prevention and treatment of hypertension.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Juan‐Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Center for Healthy AgingWuhan University School of NursingWuhanChina
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
21
|
Ye B, Fan X, Fang Z, Mao C, Lin L, Wu J, Zheng W, Cai X, Huang W, Lv Y, Han B, Han J. Macrophage-derived GSDMD promotes abdominal aortic aneurysm and aortic smooth muscle cells pyroptosis. Int Immunopharmacol 2024; 128:111554. [PMID: 38262162 DOI: 10.1016/j.intimp.2024.111554] [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: 08/14/2023] [Revised: 12/30/2023] [Accepted: 01/14/2024] [Indexed: 01/25/2024]
Abstract
Macrophage is a vital factor in determining the fate of abdominal aortic aneurysm (AAA). The crosstalk between macrophage and other cells plays a crucial role in the development of aneurysm. Gasdermin D (GSDMD) is a vital executive protein of pyroptosis, which is a novel programmed cell death associated with inflammation. In this study, we identified aortic macrophage as the main expressing cell of GSDMD in AAA. Using Gsdmd-/-ApoE-/- mouse and AAV-F4/80-shGSDMD, we demonstrated the potential role of macrophage-derived GSDMD in AAA and aortic pyroptosis induced by Ang II in vivo. In vitro experiments showed that GSDMD promotes the pyroptosis of mouse primary peritoneal macrophages (MPMs), murine aortic vascular smooth muscle cells (MOVAS) and primary smooth muscle cells. Mechanistically, a mouse cytokine antibody array showed that Gsdmd-/- inhibited LPS + nigericin (LN)- induced secretion of multiple cytokines from MPMs. Furthermore, GSDMD is involved in the crosstalk between MPMs and MOVAS via cytokine secretion. This study provides a novel fundamental insight into macrophage-derived GSDMD in AAA and showed that GSDMD could be a promising therapeutic target for AAA.
Collapse
Affiliation(s)
- Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zimin Fang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenxi Mao
- Department of Cardiac Surgery, The 1(st) Affiliated Hospital of Wenzhou Medical University, China
| | - Liming Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Wu
- Department of Vascular Surgery, The 2(nd) Affiliated Hospital of Wenzhou Medical University, China
| | - Wenyuan Zheng
- Department of Cardiology, The 1(st) Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yahui Lv
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bingjiang Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
22
|
Li K, Wei M, Zhang D, Zhai S, Liu H. PANoptosis in vascular smooth muscle cells regulated by TNF-α/IL-1β can be a new target for alleviating the progression of abdominal aortic aneurysm. Physiol Genomics 2024; 56:158-166. [PMID: 38047310 DOI: 10.1152/physiolgenomics.00053.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/15/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023] Open
Abstract
PANoptosis is an inflammatory programmed cell death (PCD) regulated by multifaceted PANoptosome complexes with major features of pyroptosis, apoptosis, and/or necroptosis that cannot be accounted for by any of these PCD pathways alone. The aim of this study was to investigate the role of PANoptosis on the occurrence and development of abdominal aortic aneurysm (AAA). Clinical samples of patients with AAA, angiotensin II (ANG II)-induced AAA mouse model, and ANG II-induced vascular smooth muscle cells (VSMCs) in vitro model were used for investigation on PANoptosis features. The expressions of ZBP1, AIM2, and other markers related to pyroptosis, apoptosis, and necroptosis elevated obviously in aortic wall tissues of patients with AAA, mice with AAA, and ANG II-treated VSMCs. ANG II treatment increased inflammatory cytokines levels in VSMCs. The stimulation of tumor necrosis factor-α (TNF-α) or interleukin-1β (IL-1β) alone promoted VSMCs death, and the effect of TNF-α combined with IL-1β is more obvious. The expressions of ZBP1, AIM2, and related markers of pyroptosis, apoptosis, and necroptosis were increased by TNF-α and IL-1β combined treatment. Inhibition of TNF-α and/or IL-1β in mice with AAA improved the AAA pathology, reduced the loss of VSMCs, decreased the expression of ZBP1 and AIM2, and markers associated with pyroptosis, apoptosis, and necroptosis. PANoptosis features were observed in aortic wall tissues of patients with AAA, mice with AAA, and ANG II-treated VSMCs. The inhibition of TNF-α and IL-1β can alleviate PANoptosis in mice with AAA, which provides a new strategy for the prevention and treatment of AAA.NEW & NOTEWORTHY Early detection, diagnosis, and treatment are very important to improve the quality of life and prognosis of patients with abdominal aortic aneurysm (AAA). Based on the findings of apoptosis, necroptosis, and pyroptosis (PANoptosis) in AAA clinical samples, this study further explored the molecular mechanism in vivo and in vitro. Specifically, inhibition of tumor necrosis factor-α and interleukin-1β can reduce PANoptosis in vascular smooth muscle cell and thus alleviate the process of AAA.
Collapse
Affiliation(s)
- Kun Li
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Henan University, Zhengzhou, People's Republic of China
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University People's Hospital, Zhengzhou, People's Republic of China
| | - Mingyang Wei
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Dongbin Zhang
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Shuiting Zhai
- Department of Vascular and Endovascular Surgery, Henan Provincial People's Hospital, People's Hospital of Henan University, Zhengzhou, People's Republic of China
| | - Hongzhi Liu
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University People's Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
23
|
Zhao S, Cang H, Liu Y, Huang Y, Zhang S. Integrated analysis of bulk RNA-seq and single-cell RNA-seq reveals the function of pyrocytosis in the pathogenesis of abdominal aortic aneurysm. Aging (Albany NY) 2023; 15:15287-15323. [PMID: 38112597 PMCID: PMC10781497 DOI: 10.18632/aging.205350] [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: 07/25/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023]
Abstract
Pyrocytosis is involved in the development of abdominal aortic aneurysm (AAA), we explored the pyrocytosis-related hub genes in AAA and conducted a diagnostic model based on the pyrocytosis-related genes score (PRGs). A total of 2 bulk RNA-seq (GSE57691 and GSE47472) datasets and pyrocytosis-related genes were integrated to obtain 24 pyrocytosis-related different expression genes (DEGs). The LASSO Cox regression analysis was conducted to filter out 7 genes and further establish the nomogram signature based on the PRGs that exhibited a good diagnosis value. Weighted gene co-expression network analysis (WGCNA) established 14 gene modules and further identified 6 hub genes which were involved in the regulatory process of pyrocytosis in AAA. At the single cell level, we further identified 3 immune cells were highly associated with the pyrocytosis process in AAA. Finally, the cell-cell communication demonstrated that fibroblasts and endothelial cells and myeloid cells maintained close communications. Here, we identified the dysfunctional expressed pyrocytosis-related genes and immune cells in AAA, which provide a comprehensive understanding of the pathogenesis of AAA.
Collapse
Affiliation(s)
- Shiqi Zhao
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Hai Cang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Ying Liu
- Department of Anesthesiology, Heilongjiang Provincial Hospital, Harbin 150036, Heilongjiang, China
| | - Yanjie Huang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Song Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| |
Collapse
|
24
|
Lu Y, Sun Y, Saaoud F, Shao Y, Xu K, Jiang X, Wu S, Yu J, Snyder NW, Yang L, Shi XM, Zhao H, Wang H, Yang X. ER stress mediates Angiotensin II-augmented innate immunity memory and facilitates distinct susceptibilities of thoracic from abdominal aorta to aneurysm development. Front Immunol 2023; 14:1268916. [PMID: 37731512 PMCID: PMC10507336 DOI: 10.3389/fimmu.2023.1268916] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023] Open
Abstract
To determine the roles of endoplasmic reticulum (ER) stress and trained immunity, we performed transcriptome analyses on the thoracic aorta (TA) and abdominal aorta (AA) from the angiotensin II (Ang II)-HFD-ApoE-KO aneurysm model and made significant findings: 1) Ang II bypassed HFD-induced metabolic reprogramming and induced stronger inflammation in AA than in TA; 2) Ang II and HFD upregulated 890 genes in AA versus TA and induced cytokine signaling; 3) Ang II AA and TA upregulated 73 and 68 cytokines, scRNA-Seq identified markers of macrophages and immune cells, cell death regulators, respectively; transdifferentiation markers of neuron, glial, and squamous epithelial cells were upregulated by Ang II-AA and TA; and pyroptosis signaling with IL-1β and caspase-4 were more upregulated in Ang II-AA than in TA; 4) Six upregulated transcriptomes in patients with AAA, Ang II AA, Ang II TA, additional aneurysm models, PPE-AAA and BAPN-Ang II-AAA, were partially overlapped with 10 lists of new ER stress gene sets including 3 interaction protein lists of ER stress regulators ATF6, PERK, and IRE1, HPA ER localization genes, KEGG signal genes, XBP1 transcription targets, ATF4 (PERK) targets, ATF6 targets, thapsigargin ER stress genes, tunicamycin-ER stress genes, respectively; 5) Ang II-AA and TA upregulated ROS regulators, MitoCarta genes, trained immunity genes, and glycolysis genes; and 6) Gene KO transcriptomes indicated that ATF6 and PERK played more significant roles than IRE1 in promoting AAA and trained immunity whereas antioxidant NRF2 inhibited them. Our unprecedented ER-focused transcriptomic analyses have provided novel insights on the roles of ER as an immune organelle in sensing various DAMPs and initiating ER stress that triggers Ang II-accelerated trained immunity and differs susceptibilities of thoracic and abdominal aortas to diseases.
Collapse
Affiliation(s)
- Yifan Lu
- Centers of Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yu Sun
- Centers of Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Fatma Saaoud
- Centers of Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Centers of Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Keman Xu
- Centers of Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Centers of Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
- Metabolic Disease Research and Thrombosis Research Center, Departments of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Sheng Wu
- Metabolic Disease Research and Thrombosis Research Center, Departments of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jun Yu
- Metabolic Disease Research and Thrombosis Research Center, Departments of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Nathaniel W. Snyder
- Metabolic Disease Research and Thrombosis Research Center, Departments of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ling Yang
- Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xinghua Mindy Shi
- Department of Computer and Information Sciences, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Huaqing Zhao
- Biomedical Education and Data Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Metabolic Disease Research and Thrombosis Research Center, Departments of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Centers of Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
- Metabolic Disease Research and Thrombosis Research Center, Departments of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
25
|
Xu J, Yang Y, Li X, Ding S, Zheng L, Xiong C, Yang Y. Pleiotropic activities of succinate: The interplay between gut microbiota and cardiovascular diseases. IMETA 2023; 2:e124. [PMID: 38867936 PMCID: PMC10989957 DOI: 10.1002/imt2.124] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/14/2024]
Abstract
Cardiovascular diseases (CVDs) continue to be a significant contributor to global mortality, imposing a substantial burden and emphasizing the urgent need for disease control to save lives and prevent disability. With advancements in technology and scientific research, novel mechanisms underlying CVDs have been uncovered, leading to the exploration of promising treatment targets aimed at reducing the global burden of the disease. One of the most intriguing findings is the relationship between CVDs and gut microbiota, challenging the traditional understanding of CVDs mechanisms and introducing the concept of the gut-heart axis. The gut microbiota, through changes in microbial compositions and functions, plays a crucial role in influencing local and systemic effects on host physiology and disease development, with its metabolites acting as key regulators. In previous studies, we have emphasized the importance of specific metabolites such as betaine, putrescine, trimethylamine oxide, and N,N,N-trimethyl-5-aminovaleric acid in the potential treatment of CVDs. Particularly noteworthy is the gut microbiota-associated metabolite succinate, which has garnered significant attention due to its involvement in various pathophysiological pathways closely related to CVDs pathogenesis, including immunoinflammatory responses, oxidative stress, and energy metabolism. Furthermore, we have identified succinate as a potential biomarker, highlighting its therapeutic feasibility in managing aortic dissection and aneurysm. This review aims to comprehensively outline the characteristics of succinate, including its biosynthetic process, summarize the current evidence linking it to CVDs causation, and emphasize the host-microbial crosstalk involved in modulating CVDs. The insights presented here offer a novel paradigm for future management and control of CVDs.
Collapse
Affiliation(s)
- Jing Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yicheng Yang
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xin Li
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shusi Ding
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain ProtectionThe Capital Medical UniversityBeijingChina
| | - Lemin Zheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain ProtectionThe Capital Medical UniversityBeijingChina
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Health Science CenterPeking UniversityBeijingChina
| | - Changming Xiong
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuejin Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
26
|
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
|