1
|
Xiao Q, Li Y, Cai B, Huang X, Fang L, Liang F, Chen L, Xu K, Zhang W, Wang X, Yin A, Wang X, Cai Z, Zhuang F, Shao Q, Zhou B, Hocher B, He B, Shen L. CCDC80 Protects against Aortic Dissection and Rupture by Maintaining the Contractile Smooth Muscle Cell Phenotype. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2502108. [PMID: 40278823 DOI: 10.1002/advs.202502108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/20/2025] [Indexed: 04/26/2025]
Abstract
Aortic dissection (AD) is a life-threatening medical emergency characterized by adverse vascular remodeling. Coiled-coil domain-containing protein 80 (CCDC80) plays an essential role in regulating cardiovascular remodeling. This study aims to define the role of CCDC80 in the formation and development of AD. Significant downregulation of CCDC80 in vascular smooth muscle cell (VSMC) in human and mouse AD is identified. Then, CCDC80 knockout mice (CCDC80-/-) and VSMC-specific CCDC80 knockout mice (CCDC80fl/fl SM22α Cre+) treated with angiotensin II (Ang II) or Ang II combined with β-aminopropionitrile monofumarate (BAPN) frequently develop AD with higher frequency and severity, accompanied by severe elastin fragmentation and collagen deposition. Mechanistically, CCDC80 interacts with JAK2, and CCDC80 deficiency promotes VSMC phenotype switching, proliferation, and migration as well as matrix metalloproteinase production by activating the JAK2/STAT3 signaling pathway. Moreover, the JAK2/STAT3 pathway-specific inhibitor ameliorates adverse vascular remodeling and reduces AD formation in CCDC80-knockout mice by mitigating VSMC phenotype switching. In conclusion, CCDC80 deficiency exacerbates the progression of events leading to AD by activating the JAK2/STAT3 pathway involved in regulating the phenotype switching and function of VSMCs. These findings highlight that CCDC80 is a potential key target for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Qingqing Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Bin Cai
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiying Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Liang Fang
- Department of Cardiac Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Long Chen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ke Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Weifeng Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xiaolei Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Fei Zhuang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Qin Shao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Bin Zhou
- Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200032, China
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, 69123, Heidelberg, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, People's Republic of China, Changsha, 410028, China
- IMD Institut fur Medizinische Diagnostik Berlin-Potsdam GbR, 14473, Berlin, Germany
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| |
Collapse
|
2
|
Lin Y, Liu Y, Feng J, Ye F, Hu L, Cao Y. Metabolomics analysis of acute lung injury induced by aortic dissection in mice. Ann Med Surg (Lond) 2025; 87:497-505. [PMID: 40110328 PMCID: PMC11918661 DOI: 10.1097/ms9.0000000000002885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 03/22/2025] Open
Abstract
Thoracic aortic aneurysm/dissection (TAA/D) is a complicated vascular disorder with galloping development and high mortality. Phenotype switching plays an important role in the pathological process of TAA/D. Previous studies indicated the potential correlation between the expression level of lncRNA RP11-465L10.10 and MMP9 involved in the development of TAA/D. Here, our results showed that RP11-465L10.10 and MMP9 were highly increased in TAD patient tissues, which was consistent in Ang II-induced vascular smooth muscle cell (VSMC) phenotype switching. However, the effects and underlying mechanism of RP11-465L10.10 on VSMC phenotypic switching remain uncertain. Therefore, the expression of SM22α, cell proliferation, and migration were investigated when ectopically expressed RP11-465L10.10 in VSMCs. The results showed that RP11-465L10.10 overexpression decreased SM22α expression and facilitated VSMC proliferation, migration, and MMP9 expression. Furthermore, the NF-κB signaling pathway was enriched in transcriptome data analysis, indicating that NF-κB signaling may be involved in RP11-465L10.10-induced VSMC phenotype switching and MMP9 expression. In conclusion, this study demonstrated that RP11-465L10.10 induces VSMC phenotype switching and MMP9 expression via the NF-κB signaling pathway, suggesting that RP11-465L10.10 might be a potential therapeutic target for TAA/D treatment.
Collapse
Affiliation(s)
- Yang Lin
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Yi Liu
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Jianfei Feng
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Fuyong Ye
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Lian Hu
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Yong Cao
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| |
Collapse
|
3
|
An Z, Sun Y, Yang X, Zhou J, Yu Y, Zhang B, Xu Z, Zhu Y, Wang G. Enhanced expression of miR-20a driven by nanog exacerbated the degradation of extracellular matrix in thoracic aortic dissection. Noncoding RNA Res 2024; 9:1040-1049. [PMID: 39022686 PMCID: PMC11254500 DOI: 10.1016/j.ncrna.2024.05.006] [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: 02/21/2024] [Revised: 05/09/2024] [Accepted: 05/19/2024] [Indexed: 07/20/2024] Open
Abstract
Thoracic aortic dissection (TAD) is a life-threatening vascular disease manifested as intramural bleeding in the medial layers of the thoracic aorta. The key histopathologic feature of TAD is medial degeneration, characterized by depletion of vascular smooth muscle cells (VSMCs) and degradation of extracellular matrix (ECM). MicroRNA, as essential epigenetic regulators, can inhibit the protein expression of target genes without modifying the sequences. This study aimed to elucidate the role and underlying mechanism of miR-20a, a member of the miR-17-92 cluster, in regulating ECM degradation during the pathogenesis of TAD. The expression of the miR-17-92 cluster was significantly increased in synthetic VSMCs derived from TAD lesions compared to contractile VSMCs isolated from normal thoracic aortas. Notably, the expression of miR-20a was increased in VSMCs in response to serum exposure and various stimuli. In TAD lesions, the expression of miR-20a was significantly negatively correlated with that of elastin. Elevated expression of miR-20a was also observed in thoracic aortas of TAD mice induced by β-aminopropionitrile fumarate and angiotensin II. Overexpression of miR-20a via mimic transfection enhanced the growth and invasive capabilities of VSMCs, with no significant impact on their migratory activity or the expression of phenotypic markers (α-SMA, SM22, and OPN). Silencing of miR-20a with inhibitor transfection mitigated the hyperactivation of MMP2 in VSMCs stimulated by PDGF-bb, as evidenced by reduced levels of active-MMP2 and increased levels of pro-MMP2. Subsequently, TIMP2 was identified as a novel target gene of miR-20a. The role of miR-20a in promoting the activation of MMP2 was mediated by the suppression of TIMP2 expression in VSMCs. In addition, the elevated expression of miR-20a was found to be directly driven by Nanog in VSMCs. Collectively, these findings indicate that miR-20a plays a crucial role in maintaining the homeostasis of the thoracic aortic wall during TAD pathogenesis and may represent a potential therapeutic target for TAD.
Collapse
Affiliation(s)
- Zhao An
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yangyong Sun
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaodong Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingwen Zhou
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yongchao Yu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Boyao Zhang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuming Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guokun Wang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
4
|
Cao X, Fang H, Zhou L. CircNRIP1 promotes proliferation, migration and phenotypic switch of Ang II-induced HA-VSMCs by increasing CXCL5 mRNA stability via recruiting IGF2BP1. Autoimmunity 2024; 57:2304820. [PMID: 38269483 DOI: 10.1080/08916934.2024.2304820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/07/2024] [Indexed: 01/26/2024]
Abstract
Circular RNA (circRNA) has been found to be differentially expressed and involved in regulating the processes of human diseases, including thoracic aortic dissection (TAD). However, the role and mechanism of circNRIP1 in the TAD process are still unclear. GEO database was used to screen the differentially expressed circRNA and mRNA in type A TAD patients and age-matched normal donors. Angiotensin II (Ang II)-induced human aortic vascular smooth muscle cells (HA-VSMCs) were used to construct TAD cell models. The expression levels of circNRIP1, NRIP1, CXC-motif chemokine 5 (CXCL5) and IGF2BP1 were detected by quantitative real-time PCR. Cell proliferation and migration were determined by EdU assay, transwell assay and wound healing assay. The protein levels of synthetic phenotype markers, contractile phenotype markers, CXCL5 and IGF2BP1 were tested by western blot analysis. The interaction between IGF2BP1 and circNRIP1/CXCL5 was confirmed by RIP assay, and CXCL5 mRNA stability was assessed by actinomycin D assay. CircNRIP1 was upregulated in TAD patients and Ang II-induced HA-VSMCs. Knockdown of circNRIP1 suppressed Ang II-induced proliferation, migration and phenotypic switch of HA-VSMCs. Also, high expression of CXCL5 was observed in TAD patients, and its knockdown could inhibit Ang II-induced HA-VSMCs proliferation, migration and phenotypic switch. Moreover, CXCL5 overexpression reversed the regulation of circNRIP1 knockdown on Ang II-induced HA-VSMCs functions. Mechanistically, circNRIP1 could competitively bind to IGF2BP1 and subsequently enhance CXCL5 mRNA stability. CircNRIP1 might contribute to TAD progression by promoting CXCL5 mRNA stability via recruiting IGF2BP1.
Collapse
Affiliation(s)
- Xianzhao Cao
- Department of Cardiothoracic Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Hongyan Fang
- Department of Emergency Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Longshu Zhou
- Department of Cardiothoracic Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
5
|
Liu Y, Yin Q, Liu B, Lu Z, Liu M, Meng L, He C, Chang J. Fisetin reduces ovalbumin-triggered airway remodeling by preventing phenotypic switching of airway smooth muscle cells. Respir Res 2024; 25:370. [PMID: 39402516 PMCID: PMC11479573 DOI: 10.1186/s12931-024-03005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The transformation of airway smooth muscle cells (ASMCs) from a quiescent phenotype to a hypersecretory and hypercontractile phenotype is a defining feature of asthmatic airway remodeling. Fisetin, a flavonoid compound, possesses anti-inflammatory characteristics in asthma; yet, its impact on airway remodeling and ASMCs phenotype transition has not been investigated. OBJECTIVES This research seeked to assess the impact of fisetin on ovalbumin (OVA) induced asthmatic airway remodeling and ASMCs phenotype transition, and clarify the mechanisms through network pharmacology predictions as well as in vivo and in vitro validation. METHODS First, a fisetin-asthma-ASMCs network was constructed to identify potential targets. Subsequently, cellular and animal studies were carried out to examine the inhibitory effects of fisetin on airway remodeling in asthmatic mice, and to detemine how fisetin impacts the phenotypic transition of ASMCs. RESULTS Network analysis indicated that fisetin might affect asthma via mediating the phosphatidylinositol 3-kinase (PI3K)/ protein kinase B (AKT) pathway. Intraperitoneal administration of fisetin in vivo reduced airway inflammation and remodeling, as shown by reduced inflammatory cells, decreased T helper type 2 (Th2) cytokine release, diminished collagen accumulation, mitigated airway smooth muscle thickening, and decreased expression of osteopontin (OPN), collagen-I and α-smooth muscle actin (α-SMA). Moreover, fisetin suppressed the PI3K/AKT pathway in asthmatic lung tissue. According to the in vitro data, fisetin downregulated the expression of the synthetic phenotypic proteins OPN and collagen-I, contractile protein α-SMA, and inhibited cellular migration, potentially through the PI3K/AKT pathway. CONCLUSION These results suggest that fisetin inhibits airway remodeling in asthma by regulating ASMCs phenotypic shift, emphasizing that fisetin is a promising candidate for the treatment of airway smooth muscle remodeling.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Qiling Yin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Bin Liu
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Zheng Lu
- Tai'an Tumour Prevention and Treatment Hospital, Tai'an, Shandong, 271000, China
| | - Meijun Liu
- Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Ling Meng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China.
| | - Chao He
- Department of Gastrointestinal Surgery, The Affiliated Taian City Central Hospital of Qingdao University, Tai'an, Shandong, 271000, China.
| | - Jin Chang
- Department of Oncology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China.
| |
Collapse
|
6
|
Zhou S, Ma B, Luo M. Matrix metalloproteinases in aortic dissection. Vascul Pharmacol 2024; 156:107420. [PMID: 39182633 DOI: 10.1016/j.vph.2024.107420] [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: 06/14/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Aortic dissection, characterized by a high immediate mortality, is primarily caused by excessive bleeding within the walls of the aorta or a severe tear within the intimal layer of the aorta. Inflammation, as well as oxidative stress and the degradation of extracellular matrix (ECM), are significant factors in the development and occurrence of aortic dissection. Matrix metalloproteinases (MMPs) are pivotal enzymes responsible for degrading the ECM. Inflammatory factors and oxidants can interact with MMPs, indicating the potential significance of MMPs in aortic dissection. A substantial body of evidence indicates that numerous MMPs are significantly upregulated in aortic dissection, playing a critical role in ECM degradation and the pathogenesis of aortic dissection. Furthermore, targeting these enzymes has demonstrated potential in facilitating ECM restoration and reducing the incidence of aortic dissection. This review initially provides a brief overview of MMP biology before delving into their expression patterns, regulatory mechanisms, and therapeutic applications in aortic dissection. A profound comprehension of the catabolic pathways associated with aortic dissection is imperative for the future development of potential preventive or therapeutic bio-interventions for aortic dissection.
Collapse
Affiliation(s)
- Shufen Zhou
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Baihui Ma
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Mingyao Luo
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Department of Vascular Surgery, Central-China Branch of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450046, China; Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650102, China.
| |
Collapse
|
7
|
Zheng L, Yang Y, Liu J, Zhao T, Zhang X, Chen L. Identification of Key Immune Infiltration Related Genes Involved in Aortic Dissection Using Bioinformatic Analyses and Experimental Verification. J Inflamm Res 2024; 17:2119-2135. [PMID: 38595338 PMCID: PMC11003470 DOI: 10.2147/jir.s434993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 03/29/2024] [Indexed: 04/11/2024] Open
Abstract
Purpose Immune microenvironment plays an important role in aortic dissection (AD). Therefore, novel immune biomarkers may facilitate AD prevention, diagnosis, and treatment. This study aimed at mining key immune-related genes and relevant mechanisms involved in AD pathogenesis. Patients and Methods Key immune cells in AD were identified by ssGESA algorithm. Next, genes associated with key immune cells were screened by weighted gene coexpression network analysis (WGCNA). Then hub immune genes were picked from protein-protein interaction network of overlapped genes from differential expression and WGCNA analyses by cytohubba plug-in. Their diagnostic potential was evaluated in two independent cohorts from GEO database. In addition, the expressions of hub immune genes were determined by quantitative RT-PCR, immunohistochemistry, and Western blotting in dissected and normal aortic tissues. Results Activated B cells, CD56dim natural killer cells, eosinophils, gamma delta T cells, immature B cells, natural killer cells and type 17 T helper cells were identified as key immune cells in AD. Thereafter, a gene module significantly correlated with key immune cells were found by WGCNA method. Subsequently, KDR, IGF1, NOS3, PECAM1, GAPDH, FLT1, DLL4, CDH5, VWF, and TEK were identified as hub immune cell related genes by PPI network analysis, which may be potential diagnostic markers for AD, as evidenced by ROC curves. Moreover, the decreased expression of VWF in AD was validated at both mRNA and protein levels, and its expression was significantly positive correlated with the marker of smooth muscle cells, ACTA2, in AD. Further immunofluorescent results showed that VWF was colocalized with ACTA2 in aortic tissues. Conclusion We identified key immune cells and hub immune cell-related genes involved in AD. Moreover, we found that VWF was co-expressed with the smooth muscle cell marker ACTA2, indicating the important role of VWF in smooth muscle cell loss in AD pathogenesis.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Vascular Surgery, the Second Hospital, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Yusi Yang
- Department of Cardiology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Jie Liu
- Department of Cardiac Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Tianliang Zhao
- Department of Cardiac Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xin Zhang
- Department of Cardiac Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Lihua Chen
- Department of Cardiac Surgery, the Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| |
Collapse
|
8
|
Gibson Hughes TA, Dona MSI, Sobey CG, Pinto AR, Drummond GR, Vinh A, Jelinic M. Aortic Cellular Heterogeneity in Health and Disease: Novel Insights Into Aortic Diseases From Single-Cell RNA Transcriptomic Data Sets. Hypertension 2024; 81:738-751. [PMID: 38318714 DOI: 10.1161/hypertensionaha.123.20597] [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: 02/07/2024]
Abstract
Aortic diseases such as atherosclerosis, aortic aneurysms, and aortic stiffening are significant complications that can have significant impact on end-stage cardiovascular disease. With limited pharmacological therapeutic strategies that target the structural changes in the aorta, surgical intervention remains the only option for some patients with these diseases. Although there have been significant contributions to our understanding of the cellular architecture of the diseased aorta, particularly in the context of atherosclerosis, furthering our insight into the cellular drivers of disease is required. The major cell types of the aorta are well defined; however, the advent of single-cell RNA sequencing provides unrivaled insights into the cellular heterogeneity of each aortic cell type and the inferred biological processes associated with each cell in health and disease. This review discusses previous concepts that have now been enhanced with recent advances made by single-cell RNA sequencing with a focus on aortic cellular heterogeneity.
Collapse
Affiliation(s)
- Tayla A Gibson Hughes
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Malathi S I Dona
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Alexander R Pinto
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| |
Collapse
|
9
|
Luo Y, Luo J, An P, Zhao Y, Zhao W, Fang Z, Xia Y, Zhu L, Xu T, Zhang X, Zhou S, Yang M, Li J, Zhu J, Liu Y, Li H, Gong M, Liu Y, Han J, Guo H, Zhang H, Jiang W, Ren F. The activator protein-1 complex governs a vascular degenerative transcriptional programme in smooth muscle cells to trigger aortic dissection and rupture. Eur Heart J 2024; 45:287-305. [PMID: 37992083 DOI: 10.1093/eurheartj/ehad534] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 07/11/2023] [Accepted: 08/09/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND AND AIMS Stanford type A aortic dissection (AD) is a degenerative aortic remodelling disease marked by an exceedingly high mortality without effective pharmacologic therapies. Smooth muscle cells (SMCs) lining tunica media adopt a range of states, and their transformation from contractile to synthetic phenotypes fundamentally triggers AD. However, the underlying pathomechanisms governing this population shift and subsequent AD, particularly at distinct disease temporal stages, remain elusive. METHODS Ascending aortas from nine patients undergoing ascending aorta replacement and five individuals undergoing heart transplantation were subjected to single-cell RNA sequencing. The pathogenic targets governing the phenotypic switch of SMCs were identified by trajectory inference, functional scoring, single-cell regulatory network inference and clustering, regulon, and interactome analyses and confirmed using human ascending aortas, primary SMCs, and a β-aminopropionitrile monofumarate-induced AD model. RESULTS The transcriptional profiles of 93 397 cells revealed a dynamic temporal-specific phenotypic transition and marked elevation of the activator protein-1 (AP-1) complex, actively enabling synthetic SMC expansion. Mechanistically, tumour necrosis factor signalling enhanced AP-1 transcriptional activity by dampening mitochondrial oxidative phosphorylation (OXPHOS). Targeting this axis with the OXPHOS enhancer coenzyme Q10 or AP-1-specific inhibitor T-5224 impedes phenotypic transition and aortic degeneration while improving survival by 42.88% (58.3%-83.3% for coenzyme Q10 treatment), 150.15% (33.3%-83.3% for 2-week T-5224), and 175.38% (33.3%-91.7% for 3-week T-5224) in the β-aminopropionitrile monofumarate-induced AD model. CONCLUSIONS This cross-sectional compendium of cellular atlas of human ascending aortas during AD progression provides previously unappreciated insights into a transcriptional programme permitting aortic degeneration, highlighting a translational proof of concept for an anti-remodelling intervention as an attractive strategy to manage temporal-specific AD by modulating the tumour necrosis factor-OXPHOS-AP-1 axis.
Collapse
Affiliation(s)
- Yongting Luo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Junjie Luo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Peng An
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Yuanfei Zhao
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Wenting Zhao
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Zhou Fang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Yi Xia
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Lin Zhu
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Teng Xu
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Xu Zhang
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Shuaishuai Zhou
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Mingyan Yang
- Analytical Biosciences Limited, Beijing 100084, China
| | - Jiayao Li
- Analytical Biosciences Limited, Beijing 100084, China
| | - Junming Zhu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Yongmin Liu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Haiyang Li
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Ming Gong
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Yuyong Liu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Jie Han
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Huiyuan Guo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| | - Hongjia Zhang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Wenjian Jiang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, No. 2 Anzhen Road, Chaoyang District, Beijing 100069, China
| | - Fazheng Ren
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, No. 10 Tianxiu Road, Haidian District, China Agricultural University, Beijing 100193, China
| |
Collapse
|
10
|
Mylonas KS, Peroulis M, Kapelouzou A. Transfection of Vein Grafts with Early Growth Response Factor-1 Oligodeoxynucleotide Decoy: Effects on Stem-Cell Genes and Toll-like Receptor-Mediated Inflammation. Int J Mol Sci 2023; 24:15866. [PMID: 37958848 PMCID: PMC10647335 DOI: 10.3390/ijms242115866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
The long-term patency of vein grafts is challenged by intimal hyperplasia. We sought to explore the intricate relationships between the transcription factor Egr-1, toll-like receptors (TLRs), and stem cell genes and also assessed oligodeoxynucleotide decoys (ODNs) as a strategy to prevent vein graft failures. A total of 42 New Zealand white rabbits were fed hyperlipidemic chow and classified into three groups. A double-stranded Egr-1 ODN was synthesized and infused in vein grafts prior to anastomosis with the common carotid artery. All vein grafts were retrieved at the conclusion of the predefined experimental period. Real-time quantitative polymerase chain reaction was performed to estimate expression patterns for several genes of interest. MYD88, TLR2-4, TLR8, NF-kB, TNF-α, IFNβ, and IFNγ; chemokines CCL4, CCL20, CCR2; numerous interleukins; and stem cell genes KFL4, NANOG, HOXA5, and HIF1α were universally downregulated in the ODN arm compared with the controls. By understanding these multifaceted interactions, our study offers actionable insights that may pave the way for innovative interventions in vascular reconstructions.
Collapse
Affiliation(s)
| | - Michail Peroulis
- Department of Surgery, Vascular Surgery Unit, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Alkistis Kapelouzou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
11
|
Sukhacheva TV, Penyaeva EV, Soborov MA, Garmanov SV, Rychin SV, Mironenko VA, Serov RA. Morphological Features of the Ascending Aorta Remodeling and Activation of Regeneratory Potential in Intima when Forming Aneurysm. Bull Exp Biol Med 2023:10.1007/s10517-023-05829-8. [PMID: 37336814 DOI: 10.1007/s10517-023-05829-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Indexed: 06/21/2023]
Abstract
In patients with an ascending aorta aneurysm, restructuring of all its layers and, first of all, the intima and media was revealed. The thickness of the intima was 79.3±63.1 μm in patients with aortic diameter <55 mm (group Ao<55) and 162.7±177.4 μm (p<0.05) in patients with aortic diameter ⩾55 mm (Ao⩾55 group), the thickness of the aortic media was 1184.0±198.2 and 1144.3±288.4 μm, respectively. In patients of the Ao<55 group, aortic dilatation was accompanied by compensatory thickening of the inner and middle layers of the aorta. In the Ao⩾55 group, thinning of the aortic media, fragmentation of elastic fibers, and its cystic degeneration were revealed. c-kit+ Stem cells were detected in the subendothelium of the thickened intima of the dilated ascending aorta. The appearance of c-kit+ cells correlated with intimal remodeling and its colonization with CD34+ and CD44+ myofibroblast-like cells.
Collapse
Affiliation(s)
- T V Sukhacheva
- A. N. Bakulev National Medical Research Center of Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - E V Penyaeva
- A. N. Bakulev National Medical Research Center of Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M A Soborov
- A. N. Bakulev National Medical Research Center of Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
- I. M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - S V Garmanov
- A. N. Bakulev National Medical Research Center of Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - S V Rychin
- A. N. Bakulev National Medical Research Center of Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V A Mironenko
- A. N. Bakulev National Medical Research Center of Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - R A Serov
- A. N. Bakulev National Medical Research Center of Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
12
|
Zhi K, Yin R, Guo H, Qu L. PUM2 regulates the formation of thoracic aortic dissection through EFEMP1. Exp Cell Res 2023; 427:113602. [PMID: 37062520 DOI: 10.1016/j.yexcr.2023.113602] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/31/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023]
Abstract
Thoracic aortic dissection (TAD) is a severe cardiovascular disease attributed to the abnormal phenotypic switch of vascular smooth muscle cells (VSMCs). We found that the RNA-binding protein PUM2 and the fibulin protein EFEMP1 were significantly decreased at the TAD anatomical site. Therefore, we constructed expression and silencing vectors for PUM2 and EFEMP1 to analyze differential expression. Overexpression of PUM2 inhibited VSMC proliferation and migration. Western blot analysis indicated that PUM2 overexpression in VSMCs upregulated α-SMA and SM22α and downregulated OPN and MMP2. Immunofluorescence demonstrated that PUM2 and EFEMP1 were co-expressed in VSMCs. Immunoprecipitation confirmed that PUM2 bound to EFEMP1 mRNA to promote EFEMP1 expression. An Ang-II-induced aortic dissection mouse model showed that PUM2 impedes the development of aortic dissection in vivo. Our study demonstrates that PUM2 inhibits the VSMC phenotypic switch to prevent aortic dissection by targeting EFEMP1 mRNA. These findings could assist the development of targeted therapy for TAD.
Collapse
Affiliation(s)
- Kangkang Zhi
- Department of Vascular Surgery, Changzheng Hospital, Second Mlitary Medical University, Shanghai, 200003, China
| | - Renqi Yin
- Department of Vascular Surgery, Changzheng Hospital, Second Mlitary Medical University, Shanghai, 200003, China
| | - Hongbo Guo
- Department of Vascular Surgery, Changzheng Hospital, Second Mlitary Medical University, Shanghai, 200003, China
| | - Lefeng Qu
- Department of Vascular Surgery, Changzheng Hospital, Second Mlitary Medical University, Shanghai, 200003, China.
| |
Collapse
|
13
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
14
|
Zhong A, Cai Y, Zhou Y, Ding N, Yang G, Chai X. Identification and Analysis of Hub Genes and Immune Cells Associated with the Formation of Acute Aortic Dissection. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2023; 2023:8072369. [PMID: 36818541 PMCID: PMC9936456 DOI: 10.1155/2023/8072369] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/19/2022] [Accepted: 01/02/2023] [Indexed: 02/10/2023]
Abstract
Background Acute type A aortic dissection (AAD) is a catastrophic disease with high mortality, but the pathogenesis has not been fully elucidated. This study is aimed at identifying hub genes and immune cells associated with the pathogenesis of AAD. Methods The datasets were downloaded from Gene Expression Omnibus (GEO). Gene Set Enrichment Analysis (GSEA), gene set variation analysis (GSVA), and differential analysis were performed. The differentially expressed genes (DEGs) were intersected with specific genes collected from MSigDB. The gene function and pathway enrichment analysis were also performed on intersecting genes. The key modules were selected by weighted gene coexpression network analysis (WGCNA). Hub genes were identified by least absolute shrinkage and selection operator (LASSO) analysis and were verified in the metadataset. The immune cell infiltration was analyzed by CIBERSORT, and the relationship between hub genes and immune cells was performed by Pearson's correlation analysis. The single-cell RNA sequencing (scRNA-seq) dataset was used to verify the differences in DNA damage and repair signaling pathways and hub genes in different cell types. Results The results of GSEA and GSVA indicated that DNA damage and repair processes were activated in the occurrence of AAD. The gene function and pathway enrichment analysis on differentially expressed DNA damage- and repair-related genes showed that these genes were mainly involved in the regulation of the cell cycle process, cellular response to DNA damage stimulus, response to wounding, p53 signaling pathway, and cellular senescence. Three key modules were identified by WGCNA. Five genes were screened as hub genes, including CDK2, EIF4A1, GLRX, NNMT, and SLCO2A1. Naive B cells and Gamma delta T cells (γδ T cells) were decreased in AAD, but monocytes and M0 macrophages were increased. scRNA-seq analysis included that DNA damage and repair processes were activated in smooth muscle cells (SMCs), tissue stem cells, and monocytes in the aortic wall of patients with AAD. Conclusions Our results suggested that DNA damage- and repair-related genes may be involved in the occurrence of AAD by regulating many biological processes. The hub genes and immune cells reported in this study also increase the understanding of AAD.
Collapse
Affiliation(s)
- Aifang Zhong
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Trauma Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuzhong Cai
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Trauma Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Trauma Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ning Ding
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Trauma Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guifang Yang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Trauma Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangping Chai
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Trauma Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Gao J, Chen Y, Wang H, Li X, Li K, Xu Y, Xie X, Guo Y, Yang N, Zhang X, Ma D, Lu HS, Shen YH, Liu Y, Zhang J, Chen YE, Daugherty A, Wang DW, Zheng L. Gasdermin D Deficiency in Vascular Smooth Muscle Cells Ameliorates Abdominal Aortic Aneurysm Through Reducing Putrescine Synthesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204038. [PMID: 36567267 PMCID: PMC9929270 DOI: 10.1002/advs.202204038] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/16/2022] [Indexed: 06/17/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a common vascular disease associated with significant phenotypic alterations in vascular smooth muscle cells (VSMCs). Gasdermin D (GSDMD) is a pore-forming effector of pyroptosis. In this study, the role of VSMC-specific GSDMD in the phenotypic alteration of VSMCs and AAA formation is determined. Single-cell transcriptome analyses reveal Gsdmd upregulation in aortic VSMCs in angiotensin (Ang) II-induced AAA. VSMC-specific Gsdmd deletion ameliorates Ang II-induced AAA in apolipoprotein E (ApoE)-/- mice. Using untargeted metabolomic analysis, it is found that putrescine is significantly reduced in the plasma and aortic tissues of VSMC-specific GSDMD deficient mice. High putrescine levels trigger a pro-inflammatory phenotype in VSMCs and increase susceptibility to Ang II-induced AAA formation in mice. In a population-based study, a high level of putrescine in plasma is associated with the risk of AAA (p < 2.2 × 10-16 ), consistent with the animal data. Mechanistically, GSDMD enhances endoplasmic reticulum stress-C/EBP homologous protein (CHOP) signaling, which in turn promotes the expression of ornithine decarboxylase 1 (ODC1), the enzyme responsible for increased putrescine levels. Treatment with the ODC1 inhibitor, difluoromethylornithine, reduces AAA formation in Ang II-infused ApoE-/- mice. The findings suggest that putrescine is a potential biomarker and target for AAA treatment.
Collapse
Affiliation(s)
- Jianing Gao
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Science of Ministry of EducationNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191P. R. China
| | - Yanghui Chen
- Division of CardiologyDepartment of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Avenue NO.1095, Qiaokou DistrictWuhan430000P. R. China
| | - Huiqing Wang
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Science of Ministry of EducationNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191P. R. China
| | - Xin Li
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Science of Ministry of EducationNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191P. R. China
| | - Ke Li
- Beijing Tiantan HospitalChina National Clinical Research Center for Neurological DiseasesAdvanced Innovation Center for Human Brain ProtectionBeijing Institute of Brain DisordersThe Capital Medical UniversityBeijing100050P. R. China
| | - Yangkai Xu
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Science of Ministry of EducationNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191P. R. China
| | - Xianwei Xie
- Department of CardiologyShengli Clinical Medical College of Fujian Medical UniversityFujian Provincial HospitalFuzhou350001P. R. China
| | - Yansong Guo
- Department of CardiologyShengli Clinical Medical College of Fujian Medical UniversityFujian Provincial HospitalFujian Provincial Key Laboratory of Cardiovascular DiseaseFujian Provincial Center for GeriatricsFujian Clinical Medical Research Center for Cardiovascular DiseasesFujian Heart Failure Center AllianceFuzhou350001P. R. China
| | - Nana Yang
- Weifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular DiseasesWeifang Medical UniversityWeifang261053P. R. China
| | - Xinhua Zhang
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyMinistry of EducationHebei Medical UniversityZhongshan East Road No. 361Shijiazhuang050017P. R. China
| | - Dong Ma
- Department of Biochemistry and Molecular BiologyThe Key Laboratory of Neural and Vascular BiologyChina Administration of EducationHebei Medical UniversityHebei050017P. R. China
| | - Hong S. Lu
- Department of PhysiologySaha Cardiovascular Research CenterUniversity of KentuckySouth LimestoneLexingtonKY40536‐0298USA
| | - Ying H. Shen
- Division of Cardiothoracic SurgeryMichael E. DeBakey Department of SurgeryBaylor College of MedicineDepartment of Cardiovascular SurgeryTexas Heart InstituteHoustonTX77030USA
| | - Yong Liu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesInstitute for Advanced StudiesWuhan UniversityWuhan430072P. R. China
| | - Jifeng Zhang
- Department of Internal MedicineUniversity of Michigan Medical CenterAnn ArborMI48109USA
| | - Y. Eugene Chen
- Department of Internal MedicineUniversity of Michigan Medical CenterAnn ArborMI48109USA
| | - Alan Daugherty
- Department of PhysiologySaha Cardiovascular Research CenterUniversity of KentuckySouth LimestoneLexingtonKY40536‐0298USA
| | - Dao Wen Wang
- Division of CardiologyDepartment of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Avenue NO.1095, Qiaokou DistrictWuhan430000P. R. China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Science of Ministry of EducationNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191P. R. China
- Beijing Tiantan HospitalChina National Clinical Research Center for Neurological DiseasesAdvanced Innovation Center for Human Brain ProtectionBeijing Institute of Brain DisordersThe Capital Medical UniversityBeijing100050P. R. China
- Hangzhou Qianjiang Distinguished ExpertHangzhou Institute of Advanced TechnologyHangzhou310026P. R. China
| |
Collapse
|
16
|
Low expression of ESR1 correlates with ascending aortic dilation and acute type A aortic dissection. Gene 2023; 851:147001. [DOI: 10.1016/j.gene.2022.147001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/03/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022]
|
17
|
Cao N, Aikeremu N, Shi WY, Tang XC, Gao RJ, Kong LJY, Zhang JR, Qin WJ, Zhang AM, Ma KT, Li L, Si JQ. Inhibition of KIR2.1 decreases pulmonary artery smooth muscle cell proliferation and migration. Int J Mol Med 2022; 50:119. [PMID: 35856410 PMCID: PMC9354699 DOI: 10.3892/ijmm.2022.5175] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022] Open
Abstract
The investigation of effective therapeutic drugs for pulmonary hypertension (PH) is critical. KIR2.1 plays crucial roles in regulating cell proliferation and migration, and vascular remodeling. However, researchers have not yet clearly determined whether KIR2.1 participates in the proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) and its role in pulmonary vascular remodeling (PVR) also remains elusive. The present study aimed to examine whether KIR2.1 alters PASMC proliferation and migration, and participates in PVR, as well as to explore its mechanisms of action. For the in vivo experiment, a PH model was established by intraperitoneally injecting Sprague-Dawley rats monocrotaline (MCT). Hematoxylin and eosin staining revealed evidence of PVR in the rats with PH. Immunofluorescence staining and western blot analysis revealed increased levels of the KIR2.1, osteopontin (OPN) and proliferating cell nuclear antigen (PCNA) proteins in pulmonary blood vessels and lung tissues following exposure to MCT, and the TGF-β1/SMAD2/3 signaling pathway was activated. For the in vitro experiments, the KIR2.1 inhibitor, ML133, or the TGF-β1/SMAD2/3 signaling pathway blocker, SB431542, were used to pre-treat human PASMCs (HPASMCs) for 24 h, and the cells were then treated with platelet-derived growth factor (PDGF)-BB for 24 h. Scratch and Transwell assays revealed that PDGF-BB promoted cell proliferation and migration. Immunofluorescence staining and western blot analysis demonstrated that PDGF-BB upregulated OPN and PCNA expression, and activated the TGF-β1/SMAD2/3 signaling pathway. ML133 reversed the proliferation and migration induced by PDGF-BB, inhibited the expression of OPN and PCNA, inhibited the TGF-β1/SMAD2/3 signaling pathway, and reduced the proliferation and migration of HPASMCs. SB431542 pre-treatment also reduced cell proliferation and migration; however, it did not affect KIR2.1 expression. On the whole, the results of the present study demonstrate that KIR2.1 regulates the TGF-β1/SMAD2/3 signaling pathway and the expression of OPN and PCNA proteins, thereby regulating the proliferation and migration of PASMCs and participating in PVR.
Collapse
Affiliation(s)
- Nan Cao
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Nigala Aikeremu
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Wen-Yan Shi
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Xue-Chun Tang
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Rui-Juan Gao
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Liang-Jing-Yuan Kong
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Jing-Rong Zhang
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Wen-Juan Qin
- Department of Ultrasound, the First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Ai-Mei Zhang
- Department of Cardiology, the First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Ke-Tao Ma
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Department of Physiology, Jiaxing University Medical College, Jiaxing, Zhejiang 314001, P.R. China
| | - Jun-Qiang Si
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
18
|
miR-335-5p regulates the proliferation, migration and phenotypic switching of vascular smooth muscle cells in aortic dissection by directly regulating SP1. Acta Biochim Biophys Sin (Shanghai) 2022; 54:961-973. [PMID: 35866606 PMCID: PMC9828317 DOI: 10.3724/abbs.2022081] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Uncontrolled proliferation, migration and phenotypic switching of vascular smooth muscle cells (VSMCs) are important steps in the development and progression of aortic dissection (AD). The function and potential mechanism of miR-335-5p in the pathogenesis of AD are explored in this study. Specifically, the biological function of miR-335-5p is explored in vitro through CCK-8, Transwell, immunofluorescence, EdU, wound-healing, RT-qPCR and western blotting assays. In addition, an AD model induced by angiotensin II is used to investigate the function of miR-335-5p in vivo. A dual-luciferase assay is performed to verify the targeting relationship between miR-335-5p and specificity protein 1 (SP1). Experiments involving the loss of SP1 function are performed to demonstrate the function of SP1 in the miR-335-5p-mediated regulation of human aortic-VSMCs (HA-VSMCs). AD tissues and platelet-derived growth factor BB (PDGF-BB)-stimulated HA-VSMCs show significant downregulation of miR-335-5p expression and upregulated SP1 expression. Overexpression of miR-335-5p effectively suppresses cell proliferation, migration and synthetic phenotype markers and enhances contractile phenotype markers induced by PDGF-BB treatment. Additionally, SP1 is identified as a target gene downstream of miR-335-5p, and its expression is negatively correlated with miR-335-5p in AD. Upregulation of SP1 partially reverses the inhibitory effect of miR-335-5p on HA-VSMCs, whereas the downregulation of SP1 has the opposite effect. Furthermore, Ad-miR-335-5p clearly suppresses aorta dilatation and vascular media degeneration in the AD model. Our results suggest that miR-335-5p inhibits HA-VSMC proliferation, migration and phenotypic switching by negatively regulating SP1, and indicate that miR-335-5p may be a potential therapeutic target in AD.
Collapse
|
19
|
Wu Y, Chen M, Chen Z, Shu J, Zhang L, Hu J, Yu H, Huang K, Liang M. Theaflavin-3,3′-Digallate from Black Tea Inhibits Neointima Formation Through Suppression of the PDGFRβ Pathway in Vascular Smooth Muscle Cells. Front Pharmacol 2022; 13:861319. [PMID: 35903325 PMCID: PMC9315285 DOI: 10.3389/fphar.2022.861319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/02/2022] [Indexed: 01/19/2023] Open
Abstract
The abnormal neointima formation caused by the phenotypic switching of vascular smooth cells (VSMCs) into a synthetic state plays a key role in the pathogenesis of various vascular diseases, including atherosclerosis and postangioplasty restenosis. Theaflavin-3,3′-digallate (TF3) in black tea has been reported to exert antiinflammatory and anticancer effects, but its role in neointima formation remains unclear. Here, we delineated a remarkable effect of TF3 in suppressing neointima formation of VSMCs in vivo as well as the ability of primary rat aortic smooth cells (RASMCs) to proliferate and migrate in vitro. Further study confirmed that the effects of TF3 on PDGF-BB–induced RASMCs were due to reduced phosphorylation of PDGFRβ, which led to the repression of downstream pathways. We concluded that TF3 may act as a repressor in the progression of neointima formation and serve as a potential therapeutic candidate for excessive phenotypic switching of VSMCs.
Collapse
Affiliation(s)
- Yichen Wu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Wuhan, China
| | - Zilong Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangcheng Shu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jiong Hu
- Department of Histology and Embryology School of Basic Medicine, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Hongjun Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Wuhan, China
- *Correspondence: Kai Huang, ; Minglu Liang,
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Wuhan, China
- *Correspondence: Kai Huang, ; Minglu Liang,
| |
Collapse
|
20
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Chen Y, Zhang T, Yao F, Gao X, Li D, Fu S, Mao L, Liu F, Zhang X, Xu Y, Deng J, Li W, Fan G, Xiao C, Chen Y, Wang L, Guo W, Zhou B. Dysregulation of interaction between LOX high fibroblast and smooth muscle cells contributes to the pathogenesis of aortic dissection. Theranostics 2022; 12:910-928. [PMID: 34976220 PMCID: PMC8692905 DOI: 10.7150/thno.66059] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/16/2021] [Indexed: 12/27/2022] Open
Abstract
Rationale: While cell-cell interaction plays a critical role in physiology and disease, a comprehensive understanding of its dynamics in vascular homeostasis and diseases is yet absent. Methods: Here, by use of single-cell RNA-sequencing and multi-color staining, we delineate the cellular composition and spatial characterization of human aorta with or without aortic dissection (AD). Results: Scrutinization of cell subtype alterations revealed significantly changed fibroblast (FB)-smooth muscle cell (SMC) interactions in AD. Of these cellular interactions, LOXhigh fibroblast (fibroblast subtype 2, FB2) in diseased state exerted the most pronounced effects on pathological deterioration of SMCs in AD. In addition, pharmacologically targeting the BMP (bone morphogenetic protein) signaling pathway effectively suppressed FB2 state transition and reduced AD incidence in mice. Finally, COL5A1 (collagen type V alpha 1 chain), one of the secreted proteins released from FB2, was significantly higher in the plasma of AD patients than in control patients, suggesting its potential use as a biomarker for AD diagnosis. Conclusions: Our work not only identified a pivotal role of a specific FB subtype in AD progression, but also shed light on cell interaction dynamics in vascular diseases.
Collapse
|
22
|
Lin Y, Huang H, Yu Y, Zhu F, Xiao W, Yang Z, Shao L, Shen Z. Long non-coding RNA RP11-465L10.10 promotes vascular smooth muscle cells phenotype switching and MMP9 expression via the NF-κB pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1776. [PMID: 35071470 PMCID: PMC8756256 DOI: 10.21037/atm-21-6402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/17/2021] [Indexed: 01/02/2023]
Abstract
Background Thoracic aortic aneurysm/dissection (TAA/D) are complicated vascular disorders with rapid development and high mortality. Vascular smooth muscle cells (VSMCs) phenotype switching plays an important role in the pathological process of TAA/D. Previous studies have indicated a potential correlation between long non-coding RNA (lncRNA) RP11-465L10.10 and matrix metallopeptidase 9 (MMP9) involved in the development of TAA/D. This study aims to investigate the role of lncRNA RP11-465L10.10 in VSMCs phenotype switching and the molecular mechanism in regulating MMP9 expression. Methods The expression of RP11-465L10.10 in vascular tissues and in VMSCs was detected by RT-qPCR. To investigate the role of RP11-465L10.10 on VSMCs phenotype switching, an RP11-465L10.10-overexpressed lentiviral vector was constructed and transfected into VSMCs. Through EdU staining, migration assay, flow cytometry analysis, the roles of RP11-465L10.10 were estimated. Bioinformatics indicated that RP11-465L10.10 upregulating MMP9 expression via NF-κB signaling, and SN50 (a specific inhibitor of NF-κB pathway) was used to inhibit the NF-κB pathway activation, then the expression of MMP9 was detected in RP11-465L10.10 overexpressed VMSCs. Results In this study, we found RP11-465L10.10 and MMP9 were highly increased in TAD patient tissues, which was consistent in angiotensin II-induced VSMCs phenotype switching. RP11-465L10.10 overexpression facilitated VSMCs phenotype switching and MMP9 expression. Mechanismly, NF-κB signal pathway was involved in RP11-465L10.10 induced VSMCs phenotype switching and MMP9 expression by transcriptome data analysis and experimental confirm. Conclusion This study demonstrated that RP11-465L10.10 induces VSMCs phenotype switching and MMP9 expression via the NF-κB signal pathway, suggesting that RP11-465L10.10 might be a potential therapeutic target for TAA/D treatment.
Collapse
Affiliation(s)
- Yang Lin
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haoyue Huang
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - You Yu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Feng Zhu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weizhang Xiao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziying Yang
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Hu C, Huang W, Xiong N, Liu X. SP1-mediated transcriptional activation of PTTG1 regulates the migration and phenotypic switching of aortic vascular smooth muscle cells in aortic dissection through MAPK signaling. Arch Biochem Biophys 2021; 711:109007. [PMID: 34400144 DOI: 10.1016/j.abb.2021.109007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 02/05/2023]
Abstract
Pituitary tumor-transforming gene 1 (PTTG1) has been found to be associated with the process of cell proliferation and invasion, and is highly expressed in aortic dissection (AD). However, its potential role and underlying mechanism in AD remain uncertain. This study aims at elucidating the roles of specificity protein 1 (SP1) and PTTG1 in the migration and phenotypic switching of aortic vascular smooth muscle cells (VSMCs) in AD. Aortic samples were collected from 35 patients with AD for examination of PTTG1 expression in the tissues by qPCR, western blot and immunofluorescence. Human aortic vascular smooth muscle cells (HAVSMCs) were stimulated with platelet-derived growth factor-BB (PDGF-BB) to establish the cellular model of AD. PTTG1 expression in VSMCs was also examined by qPCR and western blot. Cell viability was detected by CCK-8, cell proliferation by EdU staining and cell migration by wound healing and transwell. Western blot was then performed to assay migration-related proteins. After interference with PTTG1, the levels of smooth muscle pthenotypic switch markers smooth muscle protein 22 alpha (SM22-α) and osteopontin (OPN) were detected by qPCR, western blot and immunofluorescence. The binding of SP1 and PTTG1 was verified with dual-luciferase reporter assay and chromatin immunoprecipitation assay (ChIP). PTTG1 overexpression was found in AD patients. Interference with PTTG1 attenuated the proliferation and migration of PDGF-BB-stimulated HAVSMCs, in addition to their switching from contractile phenotype to synthetic phenotype. Transcription factor SP1 was up-regulated in PDGF-BB-stimulated HAVSMCs, combined with PTTG1 promoter sequence and regulated PTTG1 expression, whose overexpression reversed the effects of PTTG1 interference on cell proliferation, migration and phenotypic switching. SP1 transcriptional activation of PTTG1 activated MAPK/ERK signaling pathway. In conclusion, SP1 transcriptional activation of PTTG1 regulates the migration and phenotypic transformation of HAVSMCs in AD by MAPK Signaling.
Collapse
Affiliation(s)
- Chuangjia Hu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Weixing Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Nianling Xiong
- Shantou University Medical College, Shantou, 515000, China
| | - Xiaoqiang Liu
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China.
| |
Collapse
|
24
|
Chen Y, Wei X, Zhang Z, He Y, Huo B, Guo X, Feng X, Fang ZM, Jiang DS, Zhu XH. Downregulation of Filamin a Expression in the Aorta Is Correlated With Aortic Dissection. Front Cardiovasc Med 2021; 8:690846. [PMID: 34485398 PMCID: PMC8414519 DOI: 10.3389/fcvm.2021.690846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 07/13/2021] [Indexed: 12/23/2022] Open
Abstract
Filamins (FLNs) are actin cross-linking proteins, and as scaffolding proteins, FLNs are closely associated with the stabilization of the cytoskeleton. Nevertheless, the biological importance of FLNs in aortic dissection (AD) has not been well-elucidated. In this study, we first reanalyzed datasets downloaded from the Gene Expression Omnibus (GEO) database, and we found that in addition to the extracellular matrix, the actin cytoskeleton is a key structure associated with AD. Given that FLNs are involved in remodeling the cytoskeleton to affect cellular functions, we measured their expression levels in the aortas of patients with Stanford type A AD (TAAD). Our results showed that the mRNA and protein levels of FLNA were consistently decreased in dissected aortas of both humans and mice, while the FLNB protein level was upregulated despite decreased FLNB mRNA levels, and comparable expression levels of FLNC were observed between groups. Furthermore, the immunohistochemistry results demonstrated that FLNA was highly expressed in smooth muscle cells (SMCs) of aorta in non-AD samples, and downregulated in the medial layer of the dissected aortas of humans and mice. Moreover, we revealed that FOS and JUN, forming a dimeric transcription factor called AP-1 (activating protein-1), were positively correlated with the expression of FLNA in aorta. Either overexpression of FOS or JUN alone, or overexpression of FOS and JUN together, facilitated the expression of FLNA in primary cultured human aortic SMCs. In the present study, we not only detected the expression pattern of FLNs in aortas of humans and mice with or without AD, but we also found that the expression of FLNA in the AD samples was significantly reduced and that AP-1 might regulate the expression of FLNA. Our findings will contribute to the elucidation of the pathological mechanisms of AD and provide potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Yue Chen
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zihao Zhang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi He
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Huo
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian Guo
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Feng
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ze-Min Fang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xue-Hai Zhu
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
25
|
Campisi S, Jayendiran R, Condemi F, Viallon M, Croisille P, Avril S. Significance of Hemodynamics Biomarkers, Tissue Biomechanics and Numerical Simulations in the Pathogenesis of Ascending Thoracic Aortic Aneurysms. Curr Pharm Des 2021; 27:1890-1898. [PMID: 33319666 DOI: 10.2174/1381612826999201214231648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/06/2020] [Indexed: 11/22/2022]
Abstract
Guidelines for the treatment of aortic wall diseases are based on measurements of maximum aortic diameter. However, aortic rupture or dissections do occur for small aortic diameters. Growing scientific evidence underlines the importance of biomechanics and hemodynamics in aortic disease development and progression. Wall shear stress (WWS) is an important hemodynamics marker that depends on aortic wall morphology and on the aortic valve function. WSS could be helpful to interpret aortic wall remodeling and define personalized risk criteria. The complementarity of Computational Fluid Dynamics and 4D Magnetic Resonance Imaging as tools for WSS assessment is a promising reality. The potentiality of these innovative technologies will provide maps or atlases of hemodynamics biomarkers to predict aortic tissue dysfunction. Ongoing efforts should focus on the correlation between these non-invasive imaging biomarkers and clinico-pathologic situations for the implementation of personalized medicine in current clinical practice.
Collapse
Affiliation(s)
- Salvatore Campisi
- Department of Cardiovascular Surgery; University Hospistal of Saint Etienne, France
| | - Raja Jayendiran
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F - 42023 Saint-Etienne, France
| | - Francesca Condemi
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F - 42023 Saint-Etienne, France
| | - Magalie Viallon
- Department of Radiology, University Hospital of Saint Etienne, France
| | - Pierre Croisille
- Department of Radiology, University Hospital of Saint Etienne, France
| | - Stéphane Avril
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F - 42023 Saint-Etienne, France
| |
Collapse
|
26
|
Wang W, Liu Q, Wang Y, Piao H, Zhu Z, Li D, Wang T, Liu K. LINC01278 Sponges miR-500b-5p to Regulate the Expression of ACTG2 to Control Phenotypic Switching in Human Vascular Smooth Muscle Cells During Aortic Dissection. J Am Heart Assoc 2021; 10:e018062. [PMID: 33910387 PMCID: PMC8200748 DOI: 10.1161/jaha.120.018062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Phenotypic switching in vascular smooth muscle cells (VSMCs) is involved in the pathogenesis of aortic dissection (AD). This study aims to explore the potential mechanisms of linc01278 during VSMC phenotypic switching. Methods and Results Twelve samples (6 AD and 6 control) were used for lncRNA, microRNA, and mRNA microarray analysis. We integrated the mRNA microarray data set with GSE52093 to determine the differentially expressed genes. Bioinformatic analysis, including Gene Expression Omnibus 2R, Venn diagram analysis, gene ontology, pathway enrichment, and protein-protein interaction networks were used to identify the target lncRNA, microRNA, and mRNA involved in AD. Subsequently, we validated the bioinformatics data using techniques in molecular biology in human tissues and VSMCs. Linc01278, microRNA-500b-5p, and ACTG2 played an important role in the vascular smooth muscle contraction pathway. Linc01278 and ACTG2 were downregulated and miR-500b-5p was upregulated in AD tissues. Molecular markers of VSMC phenotypic switching, including SM22α, SMA, calponin, and MYH11, were downregulated in AD tissues. Plasmid-based overexpression and RNA interference-mediated downregulation of linc01278 weakened and enhanced VSMC proliferation and phenotypic switching, respectively. Dual-luciferase reporter assays confirmed that linc01278 regulated miR-500b-5p that directly targeted ACTG2 in HEK293T cells. Conclusions These data demonstrate that linc01278 regulates ACTG2 to control the phenotypic switch in VSMCs by sponging miR-500b-5p. This linc01278-miR-500b-5p-ACTG2 axis has a potential role in developing diagnostic markers and therapeutic targets for AD.
Collapse
Affiliation(s)
- Weitie Wang
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University Changchun Jilin China
| | - Qing Liu
- Graduate School of Medicine and Faculty of Medicine of the University of Tokyo Tokyo Japan
| | - Yong Wang
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University Changchun Jilin China
| | - Hulin Piao
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University Changchun Jilin China
| | - Zhicheng Zhu
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University Changchun Jilin China
| | - Dan Li
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University Changchun Jilin China
| | - Tiance Wang
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University Changchun Jilin China
| | - Kexiang Liu
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University Changchun Jilin China
| |
Collapse
|
27
|
Ren K, Li B, Liu Z, Xia L, Zhai M, Wei X, Duan W, Yu S. GDF11 prevents the formation of thoracic aortic dissection in mice: Promotion of contractile transition of aortic SMCs. J Cell Mol Med 2021; 25:4623-4636. [PMID: 33764670 PMCID: PMC8107100 DOI: 10.1111/jcmm.16312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/23/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Thoracic aortic dissection (TAD) is an aortic disease associated with dysregulated extracellular matrix composition and de‐differentiation of vascular smooth muscle cells (SMCs). Growth Differentiation Factor 11 (GDF11) is a member of transforming growth factor β (TGF‐β) superfamily associated with cardiovascular diseases. The present study attempted to investigate the expression of GDF11 in TAD and its effects on aortic SMC phenotype transition. GDF11 level was found lower in the ascending thoracic aortas of TAD patients than healthy aortas. The mouse model of TAD was established by β‐aminopropionitrile monofumarate (BAPN) combined with angiotensin II (Ang II). The expression of GDF11 was also decreased in thoracic aortic tissues accompanied with increased inflammation, arteriectasis and elastin degradation in TAD mice. Administration of GDF11 mitigated these aortic lesions and improved the survival rate of mice. Exogenous GDF11 and adeno‐associated virus type 2 (AAV‐2)‐mediated GDF11 overexpression increased the expression of contractile proteins including ACTA2, SM22α and myosin heavy chain 11 (MYH11) and decreased synthetic markers including osteopontin and fibronectin 1 (FN1), indicating that GDF11 might inhibit SMC phenotype transition and maintain its contractile state. Moreover, GDF11 inhibited the production of matrix metalloproteinase (MMP)‐2, 3, 9 in aortic SMCs. The canonical TGF‐β (Smad2/3) signalling was enhanced by GDF11, while its inhibition suppressed the inhibitory effects of GDF11 on SMC de‐differentiation and MMP production in vitro. Therefore, we demonstrate that GDF11 may contribute to TAD alleviation via inhibiting inflammation and MMP activity, and promoting the transition of aortic SMCs towards a contractile phenotype, which provides a therapeutic target for TAD.
Collapse
Affiliation(s)
- Kai Ren
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Buying Li
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhenhua Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lin Xia
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mengen Zhai
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xufeng Wei
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shiqiang Yu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
28
|
Cao N, Tang X, Gao R, Kong L, Zhang J, Qin W, Hu N, Zhang A, Ma K, Li L, Si JQ. Galectin-3 participates in PASMC migration and proliferation by interacting with TGF-β1. Life Sci 2021; 274:119347. [PMID: 33716065 DOI: 10.1016/j.lfs.2021.119347] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/21/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022]
Abstract
Pulmonary vascular remodelling is one of the most important factors for pulmonary hypertension (PH). Galectin-3 (Gal-3) is a β-galactoside-binding lectin. In the latest literature, Gal-3 has been reported to be involved in pulmonary vascular remodelling, and its underlying mechanism is unclear. Our research aims to prove the effect of Gal-3 on the proliferation and migration of human pulmonary artery smooth muscle cells (HPASMC) induced by transforming growth factor β1 (TGF-β1) and to study its mechanism. In vivo experiment: In Sprague-Dawley (SD) rats, monocrotaline was injected intraperitoneally to establish a PH model, and the Gal-3 inhibitor (modified citrus pectin, MCP) 28 Ds was administered in the stomach. The results indicate that Gal-3 and TGF-β1 may be involved in the occurrence and development of PH, which may be related to the Smad2/3 signalling pathway. In vitro experiment: Human pulmonary artery smooth muscle cells were pretreated with the Gal-3 inhibitor (MCP) for 24 h, then TGF-β1 or Gal-3 was administered to the cells for 24 h. The results show that exogenous TGF-β1 and Gal-3 can activate the downstream Smad2/3 signalling pathway, and increase the proliferation and migration ability of HPASMC. However, the Gal-3 inhibitor (MCP) inhibited these effects. Further results display that TGF-β1 and Gal-3 could mutually regulate the protein and mRNA expression levels. In summary, the results of this study indicate that Gal-3 regulates the Smad2/3 signalling pathway through protein interaction with TGF-β1, in turn regulates the proliferation and migration of HPASMC, thereby regulating the occurrence and development of PH.
Collapse
Affiliation(s)
- Nan Cao
- Department of Physiology, Shihezi University Medical College, Shihezi 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China
| | - Xuechun Tang
- Department of Physiology, Shihezi University Medical College, Shihezi 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China; Department of Burns department, First Affiliated Hospital of Shihezi University, Shihezi 832002, China
| | - RuiJuan Gao
- Department of Physiology, Shihezi University Medical College, Shihezi 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China
| | - Liangjingyuan Kong
- Department of Physiology, Shihezi University Medical College, Shihezi 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China
| | - Jingrong Zhang
- Department of Physiology, Shihezi University Medical College, Shihezi 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China
| | - Wenjuan Qin
- Department of Ultrasound Room, First Affiliated Hospital of Shihezi University, Shihezi 832002, China
| | - Na Hu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China
| | - Aimei Zhang
- Department of Cardiology, First Affiliated Hospital of Shihezi University, Shihezi 832002, China
| | - Ketao Ma
- Department of Physiology, Shihezi University Medical College, Shihezi 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China; Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430070, China; Department of Physiology, Huazhong University of Science and Technology of Basic Medical Sciences, Wuhan 430070, China
| | - Li Li
- Department of Physiology, Jiaxing University Medical College, Jiaxing 314001, China.
| | - Jun-Qiang Si
- Department of Physiology, Shihezi University Medical College, Shihezi 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi 832002, China; Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430070, China; Department of Physiology, Huazhong University of Science and Technology of Basic Medical Sciences, Wuhan 430070, China.
| |
Collapse
|
29
|
Han JH, Park HS, Lee DH, Jo JH, Heo KS, Myung CS. Regulation of autophagy by controlling Erk1/2 and mTOR for platelet-derived growth factor-BB-mediated vascular smooth muscle cell phenotype shift. Life Sci 2021; 267:118978. [PMID: 33412209 DOI: 10.1016/j.lfs.2020.118978] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/26/2022]
Abstract
AIMS Vascular smooth muscle cell (VSMC) phenotype shift is involved in the pathophysiology of vascular injury or platelet-derived growth factor (PDGF)-induced abnormal proliferation and migration of VSMCs. We aimed to investigate the underlying mechanism involved in PDGF-mediated signaling pathways and autophagy regulation followed by VSMC phenotype shift. MAIN METHODS The proliferation, migration and apoptosis of cultured rat aortic VSMCs were measured, and cells undergoing phenotype shift and autophagy were examined. Specific inhibitors for target proteins in signaling pathways were applied to clarify their roles in regulating cell functions. KEY FINDINGS PDGF-BB stimulation initiated autophagy activation and synthetic phenotype transition by decreasing α-smooth muscle-actin (SMA), calponin and myosin heavy chain (MHC) and increasing osteopontin (OPN) expression. However, U0126, a potent extracellular signal-regulated kinase 1/2 (Erk1/2) inhibitor, decreased PDGF-BB-induced LC3 expression, while rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR), increased it. Furthermore, U0126 decreased the expresseion of autophagy-related genes (Atgs) such as beclin-1, Atg7, Atg5, and Atg12-Atg5 complex, indicating that Erk1/2 is a regulator of PDGF-BB-induced VSMC autophagy. Regardless of autophagy inhibition by U0126 or activation by rapamycin, the PDGF-BB-induced decrease in SMA, calponin and MHC and increase in OPN expression were inhibited. Furthermore, PDGF-BB-stimulated VSMC proliferation, migration and proliferating cell nuclear antigen (PCNA) expression were inhibited by U0126 and rapamycin. SIGNIFICANCE These findings suggest that PDGF-BB-induced autophagy is strongly regulated by Erk1/2, an mTOR-independent pathway, and any approach for targeting autophagy modulation is a potential therapeutic strategy for addressing abnormal VSMC proliferation and migration.
Collapse
Affiliation(s)
- Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Republic of Korea; Institute of Drug Research & Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Republic of Korea
| | - Do-Hyung Lee
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Republic of Korea
| | - Jun-Hwan Jo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Republic of Korea
| | - Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Republic of Korea
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 34134, Republic of Korea; Institute of Drug Research & Development, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
30
|
Piao J, Park JS, Hwang DY, Hong HS, Son Y. Substance P blocks β-aminopropionitrile-induced aortic injury through modulation of M2 monocyte-skewed monocytopoiesis. Transl Res 2021; 228:76-93. [PMID: 32835906 DOI: 10.1016/j.trsl.2020.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/27/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Aortic injuries, including aortic aneurysms and dissections, are fatal vascular diseases with distinct histopathological features in the aortic tissue such as inflammation-induced endothelial dysfunction, infiltration of immune cells, and breakdown of the extracellular matrix. Few treatments are available for treating aortic aneurysms and dissections; thus, basic and clinical studies worldwide have been attempted to inhibit disease progression. Substance P (SP) exerts anti-inflammatory effects and promotes restoration of the damaged endothelium, leading to vasculature protection and facilitation of tissue repair. This study was conducted to explore the protective effects of systemically injected SP on thoracic aortic injury (TAI). A TAI animal model was induced by orally administering β-aminopropionitrile to rats for 6 weeks. β-aminopropionitrile blocked crosslinking ECM in aorta to cause structural alteration with inflammation within 1 week and then, induced aortic dissection within 4 weeks of initiating treatment, leading to mortality within 6 weeks. Treatment of TAI rats with SP-induced anti-inflammatory responses systemically and locally, possibly by enriching anti-inflammatory M2 monocytes in the spleen and peripheral blood at early phase of aortic injury due to β-aminopropionitrile. SP-induced immune suppression finally prevented the development of aortic dissection by limiting inflammation-mediated aortic destruction. Taken together, these results suggest that SP treatment can block aortic injury by controlling the immune-cell profile and suppressing proinflammatory responses during the initial stage of vascular disease progression.
Collapse
Affiliation(s)
- Jiyuan Piao
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong In, South Korea
| | - Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Dae Yeon Hwang
- East-West Medical Research Institute, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, South Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, South Korea; East-West Medical Research Institute, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, South Korea.
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong In, South Korea.
| |
Collapse
|
31
|
LncRNA H19 regulates smooth muscle cell functions and participates in the development of aortic dissection through sponging miR-193b-3p. Biosci Rep 2021; 41:227493. [PMID: 33403385 PMCID: PMC7823186 DOI: 10.1042/bsr20202298] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/03/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Multiple studies showed that long-chain noncoding RNA H19 (LncRNA H19) is high-expressed in human and mouse abdominal aortic aneurysms (AAAs). We speculated that it plays an important role in arterial disease, and therefore studied the role and mechanism of H19 in aortic dissection (AD). METHODS The expressions of related genes in human aortic smooth muscle cells (HASMCs) induced by platelet-derived growth factor BB (PDGF-BB) or in the aortic tissue of AD patients/mice were identified by Western blot and quantitative real-time polymerase chain reaction. The targeting relationship between H19 and miR-193b-3p was predicted and verified by bioinformatics analysis, dual luciferase assay, RNA pull-down assay, RNA immunoprecipitation (RIP), and Pearson correlation coefficient. The H19 and miR-193b-3p effects on the biological functions of tissues and cells were examined by MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, thiazolyl blue tetrazolium bromide) assay, wound-healing assay, and Hematoxylin-Eosin (HE) staining. RESULTS LncRNA H19 was abnormally high-expressed in thoracic aorta tissues of AD patients, and it could competitively bind to and inhibit miR-193b-3p. In the PDGF-BB group, the expressions of H19, matrix metallopeptidase (MMP) 2 (MMP-2) and MMP-9 were up-regulated and the expressions of miR-193b-3p, α-SMA, and SM22α were down-regulated; moreover, the proliferation and migration rate of HASMCs were increased. However, H19 silencing reversed the regulation of PDGF-BB on HASMCs. More interestingly, miR-193b-3p inhibitor could partially reverse the effect of H19 silencing. In addition, the above results were verified by animal experiments, showing that shH19 and up-regulated miR-193b-3p could significantly reduce the thoracic aorta pathological damage in AD mice. CONCLUSION LncRNA H19 regulated smooth muscle cell function by sponging miR-193b-3p and it participated in the development of AD.
Collapse
|
32
|
Wang W, Wang Y, Piao H, Li B, Zhu Z, Li D, Wang T, Liu K. Bioinformatics Analysis Reveals MicroRNA-193a-3p Regulates ACTG2 to Control Phenotype Switch in Human Vascular Smooth Muscle Cells. Front Genet 2021; 11:572707. [PMID: 33510768 PMCID: PMC7835941 DOI: 10.3389/fgene.2020.572707] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
Aortic dissection (AD) is among the most fatal cardiovascular diseases. However, the pathogenesis of AD remains poorly understood. This study aims to integrate the microRNAs (miRNA) and mRNA profiles and use bioinformatics analyses with techniques in molecular biology to delineate the potential mechanisms involved in the development of AD. We used the human miRNA and mRNA microarray datasets GSE98770, GSE52093, and GEO2R, Venn diagram analysis, gene ontology, and protein–protein interaction networks to identify target miRNAs and mRNAs involved in AD. RNA interference, western blotting, and luciferase reporter assays were performed to validate the candidate miRNAs and mRNAs in AD tissues and human vascular smooth muscle cells (VSMCs). Furthermore, we studied vascular smooth muscle contraction in AD. In silico analyses revealed that miR-193a-3p and ACTG2 were key players in the pathogenesis of AD. miR-193a-3p was upregulated in the AD tissues. We also found that biomarkers for the contractile phenotype in VSMCs were downregulated in AD tissues. Overexpression and depletion of miR-193a-3p enhanced and suppressed VSMC proliferation and migration, respectively. Dual luciferase reporter assays confirmed that ACTG2 was a target of miR-193a-3p. ACTG2 was also downregulated in human AD tissues and VMSCs overexpressing miR-193a-3p. Taken together, miR-193a-3p may be a novel regulator of phenotypic switching in VSMCs and the miR-193a-3p/ACTG2 axis may serve as a promising diagnostic biomarker and therapeutic candidate for AD.
Collapse
Affiliation(s)
- Weitie Wang
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| | - Yong Wang
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| | - Hulin Piao
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| | - Bo Li
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| | - Zhicheng Zhu
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| | - Dan Li
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| | - Tiance Wang
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery of the Second Hospital of Jilin University, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
33
|
Huang B, Niu Y, Chen Z, Yang Y, Wang X. Integrin α9 is involved in the pathopoiesis of acute aortic dissection via mediating phenotype switch of vascular smooth muscle cell. Biochem Biophys Res Commun 2020; 533:519-525. [PMID: 32981677 DOI: 10.1016/j.bbrc.2020.08.095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/01/2022]
Abstract
Acute aortic dissection (AAD) is a devastating disease with high mortality; however, the pathogenic mechanisms of AAD remain poorly understood. Our present study aimed to identify genes associated with AAD and explore the molecular function of candidate genes in the pathogenesis of AAD. We used a whole-genome transcriptional microarray to identify putative AAD genes using ascending aortic tissues from four patients with AAD and four healthy organ donors. The differentially expressed genes were further validated in eight patients with AAD and eight healthy organ donors. Functional assessments were conducted to analyze the effects of the identified AAD genes on the phenotype of aortic vascular smooth muscle cells (VSMCs). The whole-genome transcriptional microarray analysis found 129 dysregulated genes in the ascending aortic tissues of AAD (fold change≥2), which were mainly associated with the focal adhesion pathway and actin cytoskeleton regulation pathway. Among these genes, integrin α9 (ITGA9) was identified to be involved in both pathways and downregulated by 50% in AAD patients. The association of ITGA9 with AAD was confirmed by Western blotting analysis (P = 0.003). Functional studies showed that knocking down ITGA9 in VSMCs resulted in a decrease in contractile markers (SM22α and α-SMA) and an increase in synthetic markers (OPN and SMemb), suggesting that the VSMCs switched from a contractile to a synthetic phenotype. After overexpression of ITGA9 by a recombinant adenovirus vector in VSMCs, SM22α and α-SMA were upregulated, while SMemb was downregulated, indicating a phenotypic switch from the synthetic to contractile phenotype of VSMCs. In conclusion, our study identified ITGA9 as a novel AAD gene. This gene is downregulated in patients with AAD and is involved in the regulation of the phenotypic switch of VSMCs from a contractile to a synthetic phenotype.
Collapse
Affiliation(s)
- Bi Huang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, China; Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuting Niu
- School of Stomatology, First Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Zhaoran Chen
- Department of Geriatrics and Gerontology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanmin Yang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Xiaojian Wang
- Key Laboratory of Pulmonary Vascular Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College,Beijing, China.
| |
Collapse
|
34
|
Sawant D, Lilly B. MicroRNA-145 targets in cancer and the cardiovascular system: evidence for common signaling pathways. VASCULAR BIOLOGY 2020; 2:R115-R128. [PMID: 33283158 PMCID: PMC7709916 DOI: 10.1530/vb-20-0012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/01/2022]
Abstract
miRNAs are small regulatory RNAs which govern gene expression post-transcriptionally by primarily binding to the 3'-UTR of mRNA target genes. miR-145 is a well-studied miRNA that has been implicated in controlling a range of biological processes. miR-145 is expressed in a variety of tissues and cell types and acts as a tumor-suppressor by regulating target gene signaling pathways involved in different aspects of tumor growth and progression. There is also strong evidence that highlights the important functions of miR-145 in the cardiovascular system. Here, we review the mechanisms of miR-145 in tumorigenesis and cancer progression and compare and contrast with the roles of miR-145 in cardiovascular development and disease. We discuss the important targets of miR-145 in cancer and their possible link to the cardiovascular system.
Collapse
Affiliation(s)
- Dwitiya Sawant
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Brenda Lilly
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
35
|
Wang F, Tu Y, Gao Y, Chen H, Liu J, Zheng J. Smooth Muscle Sirtuin 1 Blocks Thoracic Aortic Aneurysm/Dissection Development in Mice. Cardiovasc Drugs Ther 2020; 34:641-650. [PMID: 32564302 DOI: 10.1007/s10557-020-07005-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE Advancing age is the major risk factor for thoracic aortic aneurysm/dissection (TAAD). However, the causative link between age-related molecules and TAAD remains elusive. Here, we investigated the role of Sirtuin 1 (SIRT1, also known as class III histone deacetylase), the best studied member of the longevity-related Sirtuin family, in TAAD development in vivo. METHODS We used male smooth muscle-specific SIRT1 transgenic (ST-Tg) mice, smooth muscle-specific SIRT1 knockout (ST-KO) mice, and their wild-type (WT) littermates on a C57BL/6J background to establish a TAAD model induced by oral administration of 3-aminopropionitrile fumarate (BAPN). We analyzed the incidence and fatality rates of TAAD in the groups. We examined matrix metallopeptidase 2 (MMP2) and MMP9 expression in aortas or cultured A7r5 cells via western blotting and real-time polymerase chain reaction (PCR). We performed chromatin immunoprecipitation (ChIP) to clarify the epigenetic mechanism of SIRT1-regulated MMP2 expression in vascular smooth muscle cells (VSMCs). RESULTS BAPN treatment markedly increased the incidence of TAAD in WT mice but caused less disease in ST-Tg mice. Moreover, ST-KO mice had the highest BAPN-induced TAAD fatality rate of all the groups. Mechanistically, SIRT1 overexpression resulted in lower MMP2 and MMP9 expression after BAPN treatment in both mouse aortas and cultured A7r5 cells. The downregulation of BAPN-induced MMP2 expression by SIRT1 was mediated by deacetylation of histone H3 lysine 9 (H3K9) on the Mmp2 promoter in the A7r5 cells. CONCLUSION Our findings suggest that SIRT1 expression in SMCs protects against TAAD and could be a novel therapeutic target for TAAD management.
Collapse
MESH Headings
- Acetylation
- Aortic Dissection/enzymology
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Animals
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Cell Line
- Disease Models, Animal
- Histones/metabolism
- Male
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Signal Transduction
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
Collapse
Affiliation(s)
- Fang Wang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yimin Tu
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Houzao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinjing Liu
- Key Laboratory of Rheumatology and Clinical Rheumatology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
36
|
Lusini M, Nenna A, Chello C, Greco SM, Gagliardi I, Nappi F, Chello M. Role of autophagy in aneurysm and dissection of the ascending aorta. Future Cardiol 2020; 16:517-526. [PMID: 32524854 DOI: 10.2217/fca-2019-0076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Maintenance of physiologically balanced levels of autophagy is crucial for cellular homeostasis and in the normal vessel wall, balanced autophagy can be considered a cytoprotective mechanism that preserves endothelial function and prevents cardiovascular disease. Recent studies pointed out the importance of the modulation of the autophagic flux in the pathogenesis of aortic dissection and aneurysms of the ascending aorta. Notably, shear stress (and its receptor p62), IL-6, Rab7 and Atg5/IRE1α pathways of autophagy may be considered the novel super-selective therapeutic target for the preventive and postoperative treatment of aortic aneurysm and aortic dissection. This review intends to summarize current evidences in this field trying to enlighten new avenues for future researches.
Collapse
Affiliation(s)
- Mario Lusini
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Antonio Nenna
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Camilla Chello
- Department of Dermatology, Università La Sapienza di Roma, Rome, Italy
| | | | - Ilaria Gagliardi
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, Paris, France
| | - Massimo Chello
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
37
|
Wang X, Zhang H, Cao L, He Y, Ma A, Guo W. The Role of Macrophages in Aortic Dissection. Front Physiol 2020; 11:54. [PMID: 32116765 PMCID: PMC7013038 DOI: 10.3389/fphys.2020.00054] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/21/2020] [Indexed: 12/21/2022] Open
Abstract
Aortic dissection (AD) is a fatal disease that accounts for a large proportion of aortic-related deaths and has an incidence of about 3–4 per 100,000 individuals every year. Recent studies have found that inflammation plays an important role in the development of AD, and that macrophages are the hub of inflammation in the aortic wall. Aortic samples from AD patients reveal a large amount of macrophage infiltration. The sites of macrophage infiltration and activity vary throughout the different stages of AD, with involvement even in the tissue repair phase of AD. Angiotensin II has been shown to be an important factor in the stimulation of macrophage activity. Stimulated macrophages can secrete metalloproteinases, inflammatory factors and other substances to cause matrix destruction, smooth muscle cell apoptosis, neovascularization and more, all of which destroy the aortic wall structure. At the same time, there are a number of factors that regulate macrophages to reduce the formation of AD and induce the repair of torn aortic tissues. The aim of this review is to take a close look at the roles of macrophages throughout the course of AD disease.
Collapse
Affiliation(s)
- Xinhao Wang
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongpeng Zhang
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Long Cao
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,Department of General Surgery, PLA No. 983 Hospital, Tianjin, China
| | - Yuan He
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Airong Ma
- Department of Obstetrics, Zibo Central Hospital, Zibo, China
| | - Wei Guo
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
38
|
Li B, Wang Z, Chen R, Hong J, Wu Q, Hu J, Hu Z, Zhang M. Up regulation of isoleucyl-tRNA synthetase promotes vascular smooth muscle cells dysfunction via p38 MAPK/PI3K signaling pathways. Life Sci 2019; 224:51-57. [DOI: 10.1016/j.lfs.2019.03.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/17/2019] [Accepted: 03/21/2019] [Indexed: 10/27/2022]
|
39
|
An Z, Qiao F, Lu Q, Ma Y, Liu Y, Lu F, Xu Z. Interleukin-6 downregulated vascular smooth muscle cell contractile proteins via ATG4B-mediated autophagy in thoracic aortic dissection. Heart Vessels 2017; 32:1523-1535. [PMID: 28965202 DOI: 10.1007/s00380-017-1054-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/22/2017] [Indexed: 01/03/2023]
Abstract
Interleukin-6 (IL-6) overexpression played an important role in the pathogenesis of thoracic aortic dissection (TAD). Our previous study found enhanced autophagy accompanying with contractile proteins α smooth muscle actin (α-SMA) and smooth muscle 22α (SM22α) degradation in TAD aortic vascular smooth muscle cells (VSMCs). Autophagy is an important way for intracellular proteins degradation, while IL-6 has been found as a contributing factor of autophagy in some cancers. These indicated IL-6 might contribute to the occurrence of TAD by promoting autophagy-induced contractile proteins degradation, which has not been investigated. The aim of the present study is to verify this hypothesis and investigate the mechanism of it. We collected 10 TAD and 10 control aortic specimens from patients underwent TAD surgical repair and coronary artery bypass grafting, respectively. Quantitative real-time polymerase chain reaction was used to detect mRNA expression. Protein expression level was assessed by enzyme-linked immunosorbent assay, western blot, and immunohistochemistry. Microtubule-associated protein 1 light chain 3 beta overexpression adenovirus with green and red fluorescent protein tags and transmission electron microscopy were used to detect autophagy level in VSMCs. 3-Methyladenine (3-MA) and chloroquine were used to block autophagy in human VSMCs. Experiment results showed that the expression of IL-6 was significantly increased accompanying with up-regulated autophagy in TAD aortic wall compared with controls. In vitro results showed that IL-6 stimulation decreased the expression of VSMCs contractile proteins α-SMA and SM22α accompanying with up-regulated autophagy. Blocking autophagy with 3-MA or chloroquine inhibited IL-6 induced α-SMA and SM22α degradation. Further investigation showed that autophagy-related 4B cysteine peptidase (ATG4B) was significantly overexpressed in TAD aortic wall and played important role in IL-6 induced autophagy up-regulation. ATG4B knockdown blocked IL-6-induced autophagy and α-SMA and SM22α degradation, while ATG4B overexpression partly replaced the function of IL-6 in human VSMCs. In conclusion, our study demonstrated that IL-6 downregulated expression of VSMCs contractile proteins α-SMA and SM22α via enhancing ATG4B-mediated autophagy in TAD.
Collapse
Affiliation(s)
- Zhao An
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Fan Qiao
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Qijue Lu
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ye Ma
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Yang Liu
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Fanglin Lu
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China.
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China.
| |
Collapse
|
40
|
Shahbazi M, Fischbein M. Novel role of NANOG in smooth muscle cell phenotypic modulation during aortic dissections. J Thorac Cardiovasc Surg 2017; 154:1522-1523. [PMID: 28826598 DOI: 10.1016/j.jtcvs.2017.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 07/14/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Mohammad Shahbazi
- Department of Cardiothoracic Surgery, Stanford University Medical Center, Stanford, Calif
| | - Michael Fischbein
- Department of Cardiothoracic Surgery, Stanford University Medical Center, Stanford, Calif.
| |
Collapse
|