1
|
Cazzola M, Calzetta L, Rogliani P, Matera MG. Emerging Anti-Inflammatory COPD Treatments: Potential Cardiovascular Impacts. Int J Chron Obstruct Pulmon Dis 2024; 19:2481-2495. [PMID: 39606712 PMCID: PMC11600434 DOI: 10.2147/copd.s498255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive inflammatory condition often complicated by cardiovascular disease (CVD) due to shared inflammatory pathways. This review explores the cardiovascular impacts of emerging anti-inflammatory therapies in COPD. Phosphodiesterase (PDE) inhibitors may offer anti-inflammatory effects with improved lung function but pose potential risks for arrhythmias when PDE3 is inhibited although PDE4 inhibitors reduce cardiovascular events by improving endothelial function and reducing thrombosis. Similarly, p38 mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) inhibitors target COPD-related inflammation and may benefit COPD patients with CVD. p38 MAPK inhibitors reduce cardiac fibrosis, enhance contractility and lower the risk of arrhythmia. PI3K inhibitors target the PI3K/Akt pathway, which drives atherosclerosis and cardiac fibrosis, and thus potentially mitigate both plaque instability and fibrosis. Biologic therapies, including monoclonal antibodies that inhibit IL-5, IL-13/IL-4, thymic stromal lymphopoietin, IL-33, and IL-17A, show promise in reducing exacerbations but require close cardiovascular monitoring due to their immunomodulatory effects. Single-target inhibitors of neutrophil elastase or matrix metalloproteinases show limited efficacy in COPD but may aid cardiovascular patients by stabilizing atherosclerotic plaques through promoting vascular smooth muscle cell proliferation. However, their tendency to degrade the extracellular matrix and attract immune cells may heighten plaque rupture risk, contraindicating use in CVD. Alpha-1 antitrypsin replacement therapy holds promise, potentially reducing COPD exacerbations and providing cardiovascular protection, especially in myocardial injury. Understanding the influence of these innovative therapies on CVD is vital, making it imperative to examine these molecules in COPD patients with CVD at an early stage.
Collapse
Affiliation(s)
- Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome ‘tor Vergata’, Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Disease and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome ‘tor Vergata’, Rome, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania ‘luigi Vanvitelli’, Naples, Italy
| |
Collapse
|
2
|
Tao Y, Li G, Yang Y, Wang Z, Wang S, Li X, Yu T, Fu X. Epigenomics in aortic dissection: From mechanism to therapeutics. Life Sci 2023; 335:122249. [PMID: 37940070 DOI: 10.1016/j.lfs.2023.122249] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Aortic dissection (AD) has an unfavorable prognosis. It requires early diagnosis, appropriate treatment strategies, and suspicion to recognize symptoms; thus, it is commonly described as an acute aortic emergency. The clinical manifestations of painless AD are complex and variable. However, there is no effective treatment to prevent the progression of AD. Therefore, study of the molecular targets and mechanisms of AD to enable prevention or early intervention is particularly important. Although multiple gene mutations have been proposed as linked to AD development, evidence that multiple epigenetic elements are strongly associated is steadily increasing. These epigenetic processes include DNA methylation, N6-methyladenosine, histone modification, non-histone posttranslational modification, and non-coding RNAs (ncRNAs). Among these processes, resveratrol targeting Sirtuin 1 (SIRT1), 5-azacytidine (5azaC) targeting DNA methyltransferase (DNMT), and vitamin C targeting ten-eleven translocation 2 (Tet2) showed unique advantages in improving AD and vascular dysfunction. Finally, we explored potential epigenetic drugs and diagnostic methods for AD, which might provide options for the future.
Collapse
Affiliation(s)
- Yan Tao
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Gang Li
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, Shandong 250021, China; Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Shizhong Wang
- The department of Cardiology surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China.
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China.
| |
Collapse
|
3
|
Li S, Li J, Cheng W, He W, Dai SS. Independent and Interactive Roles of Immunity and Metabolism in Aortic Dissection. Int J Mol Sci 2023; 24:15908. [PMID: 37958896 PMCID: PMC10647240 DOI: 10.3390/ijms242115908] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Aortic dissection (AD) is a cardiovascular disease that seriously endangers the lives of patients. The mortality rate of this disease is high, and the incidence is increasing annually, but the pathogenesis of AD is complicated. In recent years, an increasing number of studies have shown that immune cell infiltration in the media and adventitia of the aorta is a novel hallmark of AD. These cells contribute to changes in the immune microenvironment, which can affect their own metabolism and that of parenchymal cells in the aortic wall, which are essential factors that induce degeneration and remodeling of the vascular wall and play important roles in the formation and development of AD. Accordingly, this review focuses on the independent and interactive roles of immunity and metabolism in AD to provide further insights into the pathogenesis, novel ideas for diagnosis and new strategies for treatment or early prevention of AD.
Collapse
Affiliation(s)
- Siyu Li
- School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Cheng
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wenhui He
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shuang-Shuang Dai
- School of Medicine, Chongqing University, Chongqing 400044, China
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
4
|
Markin AM, Markina YV, Bogatyreva AI, Tolstik TV, Chakal DA, Breshenkov DG, Charchyan ER. The Role of Cytokines in Cholesterol Accumulation in Cells and Atherosclerosis Progression. Int J Mol Sci 2023; 24:ijms24076426. [PMID: 37047399 PMCID: PMC10094347 DOI: 10.3390/ijms24076426] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Atherosclerosis is the most common cardiovascular disease and is the number one cause of death worldwide. Today, atherosclerosis is a multifactorial chronic inflammatory disease with an autoimmune component, accompanied by the accumulation of cholesterol in the vessel wall and the formation of atherosclerotic plaques, endothelial dysfunction, and chronic inflammation. In the process of accumulation of atherogenic lipids, cells of the immune system, such as monocytes, macrophages, dendritic cells, etc., play an important role, producing and/or activating the production of various cytokines—interferons, interleukins, chemokines. In this review, we have tried to summarize the most important cytokines involved in the processes of atherogenesis.
Collapse
|
5
|
Zhang Y, Liu T, Deng Z, Fang W, Zhang X, Zhang S, Wang M, Luo S, Meng Z, Liu J, Sukhova GK, Li D, McKenzie ANJ, Libby P, Shi G, Guo J. Group 2 Innate Lymphoid Cells Protect Mice from Abdominal Aortic Aneurysm Formation via IL5 and Eosinophils. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206958. [PMID: 36592421 PMCID: PMC9982556 DOI: 10.1002/advs.202206958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Development of abdominal aortic aneurysms (AAA) enhances lesion group-2 innate lymphoid cell (ILC2) accumulation and blood IL5. ILC2 deficiency in Rorafl/fl Il7rCre/+ mice or induced ILC2 depletion in Icosfl-DTR-fl/+ Cd4Cre/+ mice expedites AAA growth, increases lesion inflammation, but leads to systemic IL5 and eosinophil (EOS) deficiency. Mechanistic studies show that ILC2 protect mice from AAA formation via IL5 and EOS. IL5 or ILC2 from wild-type (WT) mice, but not ILC2 from Il5-/- mice induces EOS differentiation in bone-marrow cells from Rorafl/fl Il7rCre/+ mice. IL5, IL13, and EOS or ILC2 from WT mice, but not ILC2 from Il5-/- and Il13-/- mice block SMC apoptosis and promote SMC proliferation. EOS but not ILC2 from WT or Il5-/- mice block endothelial cell (EC) adhesion molecule expression, angiogenesis, dendritic cell differentiation, and Ly6Chi monocyte polarization. Reconstitution of WT EOS and ILC2 but not Il5-/- ILC2 slows AAA growth in Rorafl/fl Il7rCre/+ mice by increasing systemic EOS. Besides regulating SMC pathobiology, ILC2 play an indirect role in AAA protection via the IL5 and EOS mechanism.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, Key Laboratory of Emergency and Trauma of Ministry of EducationInstitute of Cardiovascular Research of the First Affiliated HospitalHainan Medical UniversityHaikou571199China
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Tianxiao Liu
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
- Guangdong Provincial Geriatrics InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Zhiyong Deng
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
- Department of GeriatricsNational Key Clinic SpecialtyGuangzhou First People's HospitalSchool of MedicineSouth China University of TechnologyGuangzhou510180China
| | - Wenqian Fang
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
- Cardiac Regeneration and Ageing LabInstitute of Cardiovascular SciencesSchool of Life ScienceShanghai UniversityShanghai200444China
| | - Xian Zhang
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Shuya Zhang
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, Key Laboratory of Emergency and Trauma of Ministry of EducationInstitute of Cardiovascular Research of the First Affiliated HospitalHainan Medical UniversityHaikou571199China
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Minjie Wang
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Songyuan Luo
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Zhaojie Meng
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Jing Liu
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Galina K. Sukhova
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Dazhu Li
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Andrew N. J. McKenzie
- Division of Protein & Nucleic Acid ChemistryMRC Laboratory of Molecular BiologyCambridgeCB2 0QHUK
| | - Peter Libby
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Guo‐Ping Shi
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Junli Guo
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, Key Laboratory of Emergency and Trauma of Ministry of EducationInstitute of Cardiovascular Research of the First Affiliated HospitalHainan Medical UniversityHaikou571199China
| |
Collapse
|
6
|
Zhang W, He J, Liu M, Huang M, Chen Q, Dong J, Zhang H, Xie T, Yuan J, Zha L. Genetic Analysis Reveals Different Mechanisms of IL-5 Involved in the Development of CAD in a Chinese Han Population. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:1700857. [PMID: 36760349 PMCID: PMC9904894 DOI: 10.1155/2023/1700857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/13/2022] [Accepted: 11/24/2022] [Indexed: 02/04/2023]
Abstract
Background Coronary artery disease (CAD) is a complex disease and the leading cause of death worldwide. It is caused by genetic and environmental factors or their interactions. Candidate gene association studies are an important genetic strategy for the study of complex diseases, and multiple variants of inflammatory cytokines have been found to be associated with CAD using this method. Interleukin-5 (IL-5) is an important inflammatory immune response factor that plays a role in a various inflammatory disease. Clinical tests and animal experiments indicated that IL-5 is involved in CAD development, but the exact mechanisms are unclear. This study investigated the genetic relationship between the single nucleotide polymorphisms (SNPs) in IL5 and CAD. Materials and Methods Based on the Chinese Han population, we collected 1,824 patients with CAD and 1,920 control subjects and performed a two-stage case-control association analysis for three SNPs in IL5 (rs2057687, rs78546665, and rs2069812) using the high resolution melt (HRM) technology. Logistic regression analyses were applied to adjust for traditional risk factors for CAD and to perform haplotype and gene interaction analyses. Multiple linear regression analyses were used to study relationships between the selected SNPs and serum lipid levels. Results In this study, two-stage case-control association analysis revealed that the allele and genotype frequency distributions of the three IL5 SNPs were not statistically significant between the case and control groups. In addition, none of the IL5 haplotypes were associated with CAD. Further stratified analyses were conducted by sex, age, hypertension, and disease status, respectively, and the results revealed that the rs2057687 and rs2069812 of IL5 were associated with CAD in the male group (p adj = 0.025, OR, 0.77 (95% CI, 0.62-0.97); p adj = 0.016, OR, 0.82 (95% CI, 0.70-0.97), respectively); the rs2057687 and rs78546665 of IL5 were associated with late-onset CAD (p adj = 0.039, OR, 0.78 (95% CI, 0.62-0.99); p adj = 0.036, OR, 1.46 (95% CI, 1.02-1.53), respectively); the rs2069812 of IL5 was associated with CAD in the hypertension group (p adj = 0.036, OR, 0.84 (95% CI, 0.71-0.99)); and none of the SNPs in IL5 were associated with different CAD states (anatomical CAD and clinical CAD). In addition, the association between SNPs and the serum lipid levels indicated that rs78546665 was positively correlated with triglyceride levels (p = 0.012). Finally, SNP-SNP interaction analyses revealed that interactions of rs2057687 and rs2069812 were associated with CAD (p adj = 0.046, OR, 0.77 (95% CI, 0.13-4.68)). Conclusion This study suggested that the common variants of IL5 might play a role in CAD by affecting the risk factors for CAD and through SNP-SNP interactions, which provides a new target for specific treatment of CAD patients and a theoretical basis for personalized medicine.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junyi He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Meilin Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mingkai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianwen Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Maternal and Child Health Hospital, Wuhan 430070, China
| | - Jiangtao Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongsong Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tian Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingfeng Zha
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Ma M, Zhu F, Fan F, Pan J, Cao H, Zhou Q, Wang D. Relationship between preoperative lymphocyte to monocyte ratio and surgery outcome in type A aortic dissection. Front Surg 2023; 9:1103290. [PMID: 36684141 PMCID: PMC9849739 DOI: 10.3389/fsurg.2022.1103290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Background Lymphocyte-to-monocyte ratio (LMR) is associated with the mortality of cardiovascular diseases. However, the relationship between preoperative LMR and the prognosis of patients with Stanford type A aortic dissection (TAAD) undergoing surgical treatment remains to be determined. Methods We enrolled 879 patients with TAAD undergoing surgical treatment between January 2018 and December 2021. Patients were divided into two groups: the Deceased group and the Survived group. The baseline clinical and operative characteristics of the two groups were compared and analyzed. Results In univariate and multivariate logistic regression analysis, the association between LMR and in-hospital mortality was significant, and LMR (OR = 1.598, 95% CI 1.114-2.485, P = 0.022) remained significant after adjusting for confounders. Conclusion Lower LMR may be independently associated with higher in-hospital mortality in TAAD undergoing surgical treatment.
Collapse
Affiliation(s)
- Ming Ma
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China,Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Feng Zhu
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China,Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Fudong Fan
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China,Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Jun Pan
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China,Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Hailong Cao
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China,Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Qing Zhou
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China,Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China
| | - Dongjin Wang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China,Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China,Institute of Cardiothoracic Vascular Disease, Nanjing University, Nanjing, China,Correspondence: Dongjin Wang
| |
Collapse
|
8
|
Huang Z, Zhang Y, Liu R, Li Y, Rafique M, Midgley AC, Wan Y, Yan H, Si J, Wang T, Chen C, Wang P, Shafiq M, Li J, Zhao L, Kong D, Wang K. Cobalt loaded electrospun poly(ε-caprolactone) grafts promote antibacterial activity and vascular regeneration in a diabetic rat model. Biomaterials 2022; 291:121901. [DOI: 10.1016/j.biomaterials.2022.121901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
|
9
|
Li Y, Ma X, Mei S, Ji Y, Wang D, He L, Sun D, Yan J. mRNA, lncRNA, and circRNA expression profiles in a new aortic dissection murine model induced by hypoxia and Ang II. Front Cardiovasc Med 2022; 9:984087. [PMID: 36386298 PMCID: PMC9643159 DOI: 10.3389/fcvm.2022.984087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background and aims Aortic dissection (AD) is a cardiovascular emergency with degeneration of the aortic media. Mounting evidence indicates obstructive sleep apnea (OSA) as an independent risk factor for AD development with unknown mechanisms. This study aims to establish a stable murine model of OSA-related AD (OSA-AD) and uncover the potential changes in gene transcripts in OSA-AD. Materials and methods ApoE–/– mice were exposed to the chronic intermittent hypoxia (CIH) system combined with Ang II administration to establish the OSA-AD model. Pathological staining was performed to exhibit the physiological structure of the mouse aorta. The SBC mouse ceRNA microarray was used to identify significantly differentially expressed (DE) mRNAs, DE long-non-coding RNAs (DElncRNAs), and DE circular RNAs (DEcircRNAs) in OSA-AD tissues. Subsequently, bioinformatics analysis, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG), and protein–protein interaction (PPI) analyses, were performed to evaluate the function of the significantly differentially expressed transcripts (DETs). The hub genes were confirmed using quantitative real-time polymerase chain reaction (qRT-PCR). Results ApoE–/– mice exposed to CIH and Ang II showed a high ratio of aortic accident (73.33%) and significant aortic diameter dilatation (1.96 ± 0.175 mm). A total of 1,742 mRNAs, 2,625 lncRNAs, and 537 circRNAs were identified as DETs (LogFC ≥ 1.5 or ≤ –1.5, P < 0.05). GO and KEGG analyses demonstrated that the differentially expressed mRNAs (DEmRNAs) were most enriched in cell proliferation, migration, apoptosis, inflammation, and hypoxia-related terms, which are closely related to aortic structural homeostasis. The PPI network contained 609 nodes and 934 connections, the hub genes were highlighted with the CytoHubba plugin and confirmed by qRT-PCR in AD tissues. KEGG pathway analysis revealed that the cis-regulated genes of DElncRNAs and circRNAs-host genes were enriched in aortic structural homeostasis-related pathways. Conclusion Our findings help establish a de novo OSA-AD animal model using ApoE–/– mice. Many DEmRNAs, DElncRNAs, and DEcircRNAs were screened for the first time in OSA-AD tissues. Our findings provide useful bioinformatics data for understanding the molecular mechanism of OSA-AD and developing potential therapeutic strategies for OSA-AD.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhu Ma
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Mei
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueping Ji
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Dong Wang
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Liqun He
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
- Liqun He,
| | - Dating Sun
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
- Dating Sun,
| | - Jiangtao Yan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jiangtao Yan,
| |
Collapse
|
10
|
Beck-Joseph J, Tabrizian M, Lehoux S. Molecular Interactions Between Vascular Smooth Muscle Cells and Macrophages in Atherosclerosis. Front Cardiovasc Med 2021; 8:737934. [PMID: 34722670 PMCID: PMC8554018 DOI: 10.3389/fcvm.2021.737934] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is the largest contributor toward life-threatening cardiovascular events. Cellular activity and cholesterol accumulation lead to vascular remodeling and the formation of fatty plaques. Complications arise from blood clots, forming at sites of plaque development, which may detach and result in thrombotic occlusions. Vascular smooth muscle cells and macrophages play dominant roles in atherosclerosis. A firm understanding of how these cells influence and modulate each other is pivotal for a better understanding of the disease and the development of novel therapeutics. Recent studies have investigated molecular interactions between both cell types and their impact on disease progression. Here we aim to review the current knowledge. Intercellular communications through soluble factors, physical contact, and extracellular vesicles are discussed. We also present relevant background on scientific methods used to study the disease, the general pathophysiology and intracellular factors involved in phenotypic modulation of vascular smooth muscle cells. We conclude this review with a discussion of the current state, shortcomings and potential future directions of the field.
Collapse
Affiliation(s)
- Jahnic Beck-Joseph
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Stephanie Lehoux
- Department of Medicine, Lady Davis Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
11
|
Kong Y, Li HD, Wang D, Gao X, Yang C, Li M, Chang T, Liu Q. Group 2 innate lymphoid cells suppress the pathology of neuromyelitis optica spectrum disorder. FASEB J 2021; 35:e21856. [PMID: 34606651 DOI: 10.1096/fj.202100673r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 11/11/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a severe central nervous system (CNS) autoimmune disease that primarily damages the optic nerves and spinal cord. Group 2 innate lymphoid cells (ILC2) are potent producers of type 2 cytokines that orchestrate immune and inflammatory responses. However, the role of ILC2 in CNS autoimmune diseases remains unknown. In patients with NMOSD, we identified a significant reduction of ILC2 in peripheral blood, which was correlated with disease severity. Using a mouse model of NMOSD induced by intracerebral injection of NMOSD-IgG with complement, we found CNS infiltration of ILC2 mainly expressing interleukin (IL)-5 and IL-13. The depletion of ILC2 led to increased CNS lesion volume, reduced CNS glucose metabolism, and augmented astrocyte injury and demyelination. The exacerbated NMOSD pathology was accompanied by increased accumulation of Iba1+ cells and complement activity in CNS lesions. In addition, the expansion of ILC2 using IL-33 attenuated NMO pathology. Collectively, these findings suggest a beneficial role of ILC2 in NMOSD, which deserves further investigation for future design of immune therapies to treat patients with NMOSD.
Collapse
Affiliation(s)
- Ying Kong
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Han-Dong Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Dan Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaolin Gao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunsheng Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
| | - Ting Chang
- Department of Neurology, Tangdu Hospital, the Fourth Military Medical University, Xi'an, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
| |
Collapse
|
12
|
Xu JY, Xiong YY, Tang RJ, Jiang WY, Ning Y, Gong ZT, Huang PS, Chen GH, Xu J, Wu CX, Hu MJ, Xu J, Xu Y, Huang CR, Jin C, Lu XT, Qian HY, Li XD, Yang YJ. Interleukin-5-induced eosinophil population improves cardiac function after myocardial infarction. Cardiovasc Res 2021; 118:2165-2178. [PMID: 34259869 DOI: 10.1093/cvr/cvab237] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS Interleukin (IL)-5 mediates the development of eosinophils (EOS) that are essential for tissue post-injury repair. It remains unknown whether IL-5 plays a role in heart repair after myocardial infarction (MI). This study aims to test whether IL-5-induced EOS population promotes the healing and repair process post-MI and to reveal the underlying mechanisms. METHOD AND RESULTS MI was induced by permanent ligation of the left anterior descending coronary artery in wild-type C57BL/6 mice. Western blot and real-time polymerase chain reaction revealed elevated expression of IL-5 in the heart at 5 days post-MI. Immunohistostaining indicated that IL-5 was secreted mainly from macrophages and type 2 innate lymphoid cells in the setting of experimental MI. External supply of recombinant mouse IL-5 (20 min, 1 day, and 2 days after MI surgery) reduced the infarct size and increased ejection fraction and angiogenesis in the border zone. A significant expansion of EOS was detected in both the peripheral blood and infarcted myocardium after IL-5 administration. Pharmacological depletion of EOS by TRFK5 pretreatment muted the beneficial effects of IL-5 in MI mice. Mechanistic studies demonstrated that IL-5 increased the accumulation of CD206+ macrophages in infarcted myocardium at 7 days post-MI. In vitro co-culture experiments showed that EOS shifted bone marrow-derived macrophage polarization towards the CD206+ phenotypes. This activity of EOS was abolished by IL-4 neutralizing antibody, but not IL-10 or IL-13 neutralization. Western blot analyses demonstrated that EOS promoted the macrophage downstream signal transducer and activator of transcription 6 (STAT6) phosphorylation. CONCLUSION IL-5 facilitates the recovery of cardiac dysfunction post-MI by promoting EOS accumulation and subsequent CD206+ macrophage polarization via the IL-4/STAT6 axis. TRANSLATIONAL PERSPECTIVE Accumulating evidence suggests that modulation of innate and adaptive immune responses is a promising therapeutic strategy for myocardial infarction. In this study, we demonstrate that IL-5 exerts cardioprotective effects on infarcted myocardium by promoting eosinophil accumulation and subsequent CD206+ macrophage polarization via the IL-4/STAT6 axis. Hence, regulation of cardiac IL-5 level or eosinophil count may become a therapeutic approach for post-myocardial infarction cardiac repair in humans.
Collapse
Affiliation(s)
- Jun-Yan Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Rui-Jie Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Wen-Yang Jiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Yu Ning
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Pei-Sen Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China.,Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Gui-Hao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Jun Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Chun-Xiao Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Meng-Jin Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Yi Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Cun-Rong Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Chen Jin
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Xiao-Tong Lu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Hai-Yan Qian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Xiang-Dong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Bei Li Shi Road, Xicheng District, Beijing, 100037, People's Republic of China
| |
Collapse
|
13
|
Wang P, Wang Z, Zhang M, Wu Q, Shi F. Lnc-OIP5-AS1 exacerbates aorta wall injury during the development of aortic dissection through upregulating TUB via sponging miR-143-3p. Life Sci 2021; 271:119199. [PMID: 33577845 DOI: 10.1016/j.lfs.2021.119199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 01/01/2023]
Abstract
AIMS Dysfunction of major cells constituting the aortic wall is the pathological basis for AD development. Determining whether non-coding RNAs can influence AD progression by regulating these cellular functions and identifying some specific non-coding RNAs is of great significance in uncovering molecular mechanisms of the development of AD. MAIN METHODS Microarray analyses and hierarchical clustering analysis were used to select candidate lncRNAs and miRNAs associated with AD. Dual-luciferase reporter assay, RNA immunoprecipitation, and RNA pull-down assay were performed to verify the direct bonding relationship between genes. The regulatory effects of genes on cell function were examined in a series of experiments. KEY FINDINGS We found that lnc-OIP5-AS1 was upregulated, whereas miR-143-3p was downregulated in cells treated with angiotensin II (AngII) and AD tissues. Lnc-OIP5-AS1 functioned as a competing endogenous RNA (ceRNA) of miR-143-3p to suppress the proliferation and mobility, but promote apoptosis of HAECs and HASMCs, and simultaneously result in the imbalances between MMP-2/9 and TIMP-2/1 in HASMCs and the excessive secretion of IL-6, IL-1β, and IL-17A of HAAFs. Moreover, overexpression or silence of TUB, a target gene of miR-143-3p, counteracted the influence of miR-143-3p or lnc-OIP5-AS1 on cells, respectively. SIGNIFICANCE Our findings revealed that lncRNA OIP5-AS1 exacerbates aorta intima, media, and adventitia injury in the development of AD through upregulating TUB via sponging miR-143-3p and also support more detailed future studies by providing a novel molecular basis underlying AD formation.
Collapse
Affiliation(s)
- Peng Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan 430000, Hubei Province, People's Republic of China; Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China; Central Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan 430000, Hubei Province, People's Republic of China; Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China.
| | - Min Zhang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan 430000, Hubei Province, People's Republic of China; Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China.
| | - Qi Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan 430000, Hubei Province, People's Republic of China; Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China; Central Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China
| | - Feng Shi
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan 430000, Hubei Province, People's Republic of China; Cardiovascular Surgery Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China; Central Laboratory, Renmin Hospital of Wuhan University, 9# Zhangzhidong Road, Wuhan 430000, Hubei Province, People's Republic of China
| |
Collapse
|
14
|
ALKBH5 Exacerbates Aortic Dissection by Promoting Inflammatory Response and Apoptosis of Aortic Smooth Muscle Cells via Regulating lnc-TMPO-AS1/EZH2/IRAK4 Signals in an m6A Modification Manner. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021. [DOI: 10.1155/2021/5513966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Recently, mounting evidence indicates that N6-methyladenosine (m6A) modification functions as a pivotal posttranscriptional modification that regulates noncoding RNA biogenesis to influence the progression of multiple diseases. However, whether m6A modification is involved in aortic dissection (AD) development has never been reported. Meanwhile, numerous studies have shown that AngII-induced inflammatory damage and excessive apoptosis of human aortic smooth muscle cells (HASMCs) are the crucial pathological features of AD development. Therefore, in this study, we intended to explore whether m6A modification can regulate AD progression by influencing the damage effects of AngII on HASMCs and elucidate the underlying mechanisms. Firstly, we screened and confirmed the high expression of alkylation repair homolog protein 5 (ALKBH5), a key m6A demethylase, in aortic tissues from AD patients, indicating that m6A modification may indeed be involved in AD progression. Subsequently, we demonstrated that ALKBH5 can exacerbate the AngII-induced HASMC inflammatory injury as well as apoptosis and shorten the survival time of AngII-infused mice. Mechanistically, we revealed that lncRNA TMPO-AS1 is a downstream target for ALKBH5 to affect AD progression in vitro and vivo. Meanwhile, we confirmed that ALKBH5-mediated m6A demethylation downregulates lnc-TMPO-AS1 by decreasing the stability of its nascent. Further, we demonstrated that lnc-TMPO-AS1 exhibits its functions in HASMCs, at least partly, through downregulating IRAK4 at the epigenetic level by combining with EZH2. Finally, the direct positive correlation between ALKBH5 and IRAK4 in terms of the expression level and biological function was confirmed, which further enforced the preciseness and correctness of our findings. In conclusion, our study demonstrated that ALKBH5 aggravates AD by promoting inflammatory response and apoptosis of HASMCs via regulating lnc-TMPO-AS1/EZH2/IRAK4 signals in an m6A modification manner and may provide a novel molecular basis for subsequent researchers to searching for novel therapeutic approaches to improve the health of patients fighting AD and other cardiovascular diseases.
Collapse
|