101
|
Han J, Tan C, Pan Y, Qu C, Wang Z, Wang S, Wang C, Xu K. Andrographolide inhibits the proliferation and migration of vascular smooth muscle cells via PI3K/AKT signaling pathway and amino acid metabolism to prevent intimal hyperplasia. Eur J Pharmacol 2023; 959:176082. [PMID: 37783303 DOI: 10.1016/j.ejphar.2023.176082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/04/2023]
Abstract
Andrographolide (AGP) exerts pharmacological effects when used for the treatment of cardiovascular disease, but the molecular mechanisms underlying its inhibitory effects on the proliferation and migration of vascular smooth muscle cells (VSMCs) and intimal hyperplasia (IH) are unknown. The proliferation and migration of VSMCs treated with AGP were examined using the CCK-8, flow cytometry, and wound healing assays. Expression levels of proteins related to cell proliferation and apoptosis were quantified. Multi-omics analysis with RNA-seq and metabolome was used to explore the potential molecular mechanism of AGP treatment. Additionally, an in vivo model was established through ligation of the left common carotid artery to identify the therapeutic potential of AGP in IH. Molecular docking and western blotting were performed to verify the mechanism discovered with multi-omics analysis. The results showed that AGP inhibited the proliferation and migration of cultured VSMCs in a dose-dependent manner and alleviated IH-related vascular stenosis. AGP significantly downregulated the protein levels of CDK1, CCND1, and BCL2 and upregulated the protein level of BAX. Gene expression profiles showed a total of 3,298 differentially expressed genes (DEGs) after AGP treatment, of which 1,709 DEGs had upregulated expression and 1,589 DEGs had downregulated expression. KEGG enrichment analysis highlighted the PI3K/AKT signaling pathway, verified with the detection of the activation of PI3K and AKT phosphorylation. Further GO enrichment combined with metabolomics analysis showed that AGP inhibition in cultured VSMCs involved the amino acid metabolic process, and the expression levels of the two key factors PRDM16 and EZH2, identified with PPI and docking analysis, were significantly inhibited by AGP treatment. In conclusion, our study showed that AGP inhibited VSMCs proliferation and migration by suppressing the PI3K/AKT signaling pathway and amino acid metabolism, which, in turn, improved IH.
Collapse
Affiliation(s)
- Juanjuan Han
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chunmei Tan
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yijing Pan
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chuang Qu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Zijun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Shunshun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chunli Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Kang Xu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
102
|
Chen Y, He S, Zeng A, He S, Jin X, Li C, Mei W, Lu Q. Inhibitory Effect of β-Sitosterol on the Ang II-Induced Proliferation of A7r5 Aortic Smooth Muscle Cells. Anal Cell Pathol (Amst) 2023; 2023:2677020. [PMID: 38028434 PMCID: PMC10645495 DOI: 10.1155/2023/2677020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/01/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Objective To explore the effects of β-sitosterol on VSMC proliferation. Materials and Methods A7r5 cells were pretreated with 2 µM angiotensin II (Ang II) for 24 hr to establish an excessive VSMC proliferation model, followed by treatment with β-sitosterol for 24 hr. Cells were divided into five groups: control, Ang II, and Ang II + β-sitosterol (2, 4, 8 µM). CCK-8 assay, flow cytometry, and Ad-mCherry-GFP-LC3B assay analyzed cell proliferation, cell cycle, cell apoptosis, and autophagic flux. Additionally, the expression of proteins was detected by the western blotting. Results β-Sitosterol effectively inhibited Ang II-induced A7r5 cell proliferation (IC50 : 6.841 µM at 24 hr). It achieved this by arresting cell cycle progression, promoting apoptosis, inhibiting autophagy, and suppressing the contractile-synthetic phenotypic switch. Mechanistically, β-sitosterol downregulated PCNA, Cyclin D1, and Bcl-2, while upregulating pro-caspase 3, cleaved-caspase 3, and Bax to induce cell cycle arrest and apoptosis. Additionally, it suppressed the contractile-synthetic phenotypic transformation by downregulating OPN and upregulating α-SMA. The Ad-mCherry-GFP-LC3B Assay and western blotting revealed β-sitosterol's autophagy inhibitory effects by downregulating LC3, ULK1, and Beclin-1 while upregulating P62 expression. Discussion and Conclusion. This study found for the first time that β-sitosterol could inhibit the proliferation of A7r5 cells induced by Ang II. β-Sitosterol treatment may be recommended as a therapeutic strategy to prevent the cardiovascular diseases.
Collapse
Affiliation(s)
- Yuankun Chen
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shumiao He
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ao Zeng
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
| | - Siqing He
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chunmei Li
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China
| | - Wenjie Mei
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qun Lu
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280 East Outer Ring Road, Panyu District, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Province Engineering and Technology Center for Molecular Probe and Bio-medicine Imaging, Guangzhou, China
| |
Collapse
|
103
|
Ouyang J, Wang H, Huang J. The role of lactate in cardiovascular diseases. Cell Commun Signal 2023; 21:317. [PMID: 37924124 PMCID: PMC10623854 DOI: 10.1186/s12964-023-01350-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/06/2023] [Indexed: 11/06/2023] Open
Abstract
Cardiovascular diseases pose a major threat worldwide. Common cardiovascular diseases include acute myocardial infarction (AMI), heart failure, atrial fibrillation (AF) and atherosclerosis. Glycolysis process often has changed during these cardiovascular diseases. Lactate, the end-product of glycolysis, has been overlooked in the past but has gradually been identified to play major biological functions in recent years. Similarly, the role of lactate in cardiovascular disease is gradually being recognized. Targeting lactate production, regulating lactate transport, and modulating circulating lactate levels may serve as potential strategies for the treatment of cardiovascular diseases in the future. The purpose of this review is to integrate relevant clinical and basic research on the role of lactate in the pathophysiological process of cardiovascular disease in recent years to clarify the important role of lactate in cardiovascular disease and to guide further studies exploring the role of lactate in cardiovascular and other diseases. Video Abstract.
Collapse
Affiliation(s)
- Jun Ouyang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hui Wang
- School of Pharmacy, Guangxi Medical University, Nanning, China.
| | - Jiangnan Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
104
|
González-Salvatierra S, García-Fontana C, Lacal J, Andújar-Vera F, Martínez-Heredia L, Sanabria-de la Torre R, Ferrer-Millán M, Moratalla-Aranda E, Muñoz-Torres M, García-Fontana B. Cardioprotective function of sclerostin by reducing calcium deposition, proliferation, and apoptosis in human vascular smooth muscle cells. Cardiovasc Diabetol 2023; 22:301. [PMID: 37919715 PMCID: PMC10623848 DOI: 10.1186/s12933-023-02043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Sclerostin is an inhibitor of the Wnt/b-catenin pathway, which regulates bone formation, and can be expressed in vascular smooth muscle cells (VSMCs). Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular disease (CVD) and increased serum and tissue expression of sclerostin. However, whether the role of sclerostin is detrimental or protective in the development of CVD is unknown. Therefore, our aims are to determine the level of sclerostin in T2D patients with/without CVD and in controls, both at serum and vascular tissue, and to analyze the role of sclerostin in VSMCs under calcified environments. METHODS Cross-sectional study including 121 controls and 139 T2D patients with/without CVD (48/91). Sclerostin levels in serum were determined by ELISA, and sclerostin expression was analyzed by RT-qPCR and immunohistochemistry in calcified and non-calcified artery of lower limb from T2D patients (n = 7) and controls (n = 3). In vitro experiments were performed in VSMCs (mock and sclerostin overexpression) under calcifying conditions analyzing the sclerostin function by determination of calcium and phosphate concentrations, and quantification of calcium deposits by Alizarin Red. Proliferation and apoptosis were analyzed by MTT assay and flow cytometry, respectively. The regulation of the expression of genes involved in bone metabolism was determined by RT-qPCR. RESULTS A significant increase in serum sclerostin levels in T2D patients with CVD compared to T2D patients without CVD and controls (p < 0.001) was observed. Moreover, higher circulating sclerostin levels were independently associated with CVD in T2D patients. Increased sclerostin expression was observed in calcified arteries of T2D patients compared to non-calcified arteries of controls (p = 0.003). In vitro experiments using VSMCs under calcified conditions, revealed that sclerostin overexpression reduced intracellular calcium (p = 0.001), calcium deposits (p < 0.001), cell proliferation (p < 0.001) and promoted cell survival (p = 0.015). Furthermore, sclerostin overexpression exhibited up-regulation of ALPL (p = 0.009), RUNX2 (p = 0.001) and COX2 (p = 0.003) and down-regulation of inflammatory genes, such as, IL1β (p = 0.005), IL6 (p = 0.001) and IL8 (p = 0.003). CONCLUSIONS Sclerostin could play a protective role in the development of atherosclerosis in T2D patients by reducing calcium deposits, decreasing proliferation and inflammation, and promoting cell survival in VSMCs under calcifying conditions. Therefore, considering the bone-vascular axis, treatment with anti-sclerostin for bone disease should be used with caution.
Collapse
Affiliation(s)
- Sheila González-Salvatierra
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, 18071, Spain
| | - Cristina García-Fontana
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain.
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain.
- Institute of Health Carlos III, CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, 28029, Spain.
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Francisco Andújar-Vera
- Bioinformatic Research Service, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Department of Computer Science and Artificial Intelligence, University of Granada, Granada, 18071, Spain
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), Granada, 18014, Spain
| | | | - Raquel Sanabria-de la Torre
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Department of Biochemistry, Molecular Biology III and Immunology, University of Granada, Granada, 18071, Spain
| | - María Ferrer-Millán
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
| | - Enrique Moratalla-Aranda
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Nuclear Medicine Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain
| | - Manuel Muñoz-Torres
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain.
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain.
- Institute of Health Carlos III, CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, 28029, Spain.
- Department of Medicine, University of Granada, Granada, 18016, Spain.
| | - Beatriz García-Fontana
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Institute of Health Carlos III, CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, 28029, Spain
- Department of Cell Biology, University of Granada, Granada, 18016, Spain
| |
Collapse
|
105
|
Kawaguchi Y, Kuwana M. Pathogenesis of vasculopathy in systemic sclerosis and its contribution to fibrosis. Curr Opin Rheumatol 2023; 35:309-316. [PMID: 37490353 DOI: 10.1097/bor.0000000000000959] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
PURPOSE OF REVIEW In patients with systemic sclerosis (SSc), vascular manifestations precede skin and organ fibrosis. There is increasing evidence demonstrating a pathogenic link between early vascular injury and subsequent development of tissue fibrosis. RECENT FINDINGS Our knowledge of cellular and molecular mechanisms underlying a unique relationship between SSc-related vasculopathy and fibrosis has changed over the last few years. There is increasing evidence showing viral infection as a potential trigger elucidating vascular injury. Due to defective vascular repair machinery, this initial event results in endothelial cell activation and apoptosis as well as the recruitment of inflammatory/immune cells, leading to endothelial-to-mesenchymal transition. This sequential process induces destructive vasculopathy in capillaries, fibroproliferative vascular lesions in arteries, and excessive fibrosis in the surrounding tissue. A variety of molecular mechanisms and pathways involved in vascular remodeling linked to subsequent excessive fibrosis have been identified and serve as attractive therapeutic targets for SSc. SUMMARY Endothelial injury may play a central role in connecting three features that characterize SSc pathogenesis: vasculopathy, chronic inflammation, and fibrosis. Our understanding of the processes responsible for myofibroblast differentiation triggered by vascular injury will provide the rationale for novel targeted therapies for SSc.
Collapse
Affiliation(s)
- Yasushi Kawaguchi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
106
|
Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, Liu J. Vascular calcification: from the perspective of crosstalk. MOLECULAR BIOMEDICINE 2023; 4:35. [PMID: 37851172 PMCID: PMC10584806 DOI: 10.1186/s43556-023-00146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
Vascular calcification (VC) is highly correlated with cardiovascular disease morbidity and mortality, but anti-VC treatment remains an area to be tackled due to the ill-defined molecular mechanisms. Regardless of the type of VC, it does not depend on a single cell but involves multi-cells/organs to form a complex cellular communication network through the vascular microenvironment to participate in the occurrence and development of VC. Therefore, focusing only on the direct effect of pathological factors on vascular smooth muscle cells (VSMCs) tends to overlook the combined effect of other cells and VSMCs, including VSMCs-VSMCs, ECs-VMSCs, Macrophages-VSMCs, etc. Extracellular vesicles (EVs) are a collective term for tiny vesicles with a membrane structure that are actively secreted by cells, and almost all cells secrete EVs. EVs docked on the surface of receptor cells can directly mediate signal transduction or transfer their contents into the cell to elicit a functional response from the receptor cells. They have been proven to participate in the VC process and have also shown attractive therapeutic prospects. Based on the advantages of EVs and the ability to be detected in body fluids, they may become a novel therapeutic agent, drug delivery vehicle, diagnostic and prognostic biomarker, and potential therapeutic target in the future. This review focuses on the new insight into VC molecular mechanisms from the perspective of crosstalk, summarizes how multi-cells/organs interactions communicate via EVs to regulate VC and the emerging potential of EVs as therapeutic methods in VC. We also summarize preclinical experiments on crosstalk-based and the current state of clinical studies on VC-related measures.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qian Chen
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Xie
- Department of Orthopaedics, Movement System Injury and Repair Research Centre, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
107
|
Abstract
The medial layer of the arterial wall is composed mainly of vascular smooth muscle cells (VSMCs). Under physiological conditions, VSMCs assume a contractile phenotype, and their primary function is to regulate vascular tone. In contrast with terminally differentiated cells, VSMCs possess phenotypic plasticity, capable of transitioning into other cellular phenotypes in response to changes in the vascular environment. Recent research has shown that VSMC phenotypic switching participates in the pathogenesis of atherosclerosis, where the various types of dedifferentiated VSMCs accumulate in the atherosclerotic lesion and participate in the associated vascular remodeling by secreting extracellular matrix proteins and proteases. This review article discusses the 9 VSMC phenotypes that have been reported in atherosclerotic lesions and classifies them into differentiated VSMCs, intermediately dedifferentiated VSMCs, and dedifferentiated VSMCs. It also provides an overview of several methodologies that have been developed for studying VSMC phenotypic switching and discusses their respective advantages and limitations.
Collapse
Affiliation(s)
- Runji Chen
- Shantou University Medical CollegeShantouChina
| | - David G. McVey
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
| | - Daifei Shen
- Research Center for Translational MedicineThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | | | - Shu Ye
- Shantou University Medical CollegeShantouChina
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
- Cardiovascular‐Metabolic Disease Translational Research ProgrammeNational University of SingaporeSingapore
| |
Collapse
|
108
|
Ma W, Wei D, Li X, Shan L, Fan H, Jin H, Song B, Zhang B. CircPCNX Promotes PDGF-BB-Induced Proliferation and Migration of Human Aortic Vascular Smooth Muscle Cells Through Regulating miR-1278/DNMT1 Axis. Cardiovasc Drugs Ther 2023; 37:877-889. [PMID: 35670983 DOI: 10.1007/s10557-022-07342-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Human aortic vascular smooth muscle cells (HA-VSMCs) play vital roles in the pathogenesis of vascular diseases. Circular RNAs (circRNAs) have been reported to regulate the biological functions of HA-VSMCs. In this study, the functions of circRNA pecanex homolog (circPCNX) in platelet-derived growth factor-BB (PDGF-BB)-induced HA-VSMCs were investigated. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to determine the expression of circPCNX, DNA methyltransferase 1 (DNMT1), and microRNA-1278 (miR-1278). 5'-Ethynyl-2'-deoxyuridine (EdU) assay, flow cytometry analysis, wound healing assay, and transwell assay were used to examine cell proliferation, cell cycle, and migration. Western blot assay was utilized to measure protein levels. RNA immunoprecipitation (RIP) assay, RNA pull down assay, and dual-luciferase reporter assay were adopted to analyze the relationships among circPCNX, miR-1278, and DNMT1. RESULTS CircPCNX was upregulated in PDGF-BB-treated HA-VSMCs in a dose- or time-dependent manner. CircPCNX knockdown alleviated PDGF-BB-induced cell proliferation, cell cycle progression, and migration in HA-VSMCs. CircPCNX knockdown could reverse PDGF-BB-induced HA-VSMC progression by regulating DNMT1. Moreover, circPCNX was identified to regulate DNMT1 expression by sponging miR-1278. Inhibition of miR-1278 reversed circPCNX knockdown-mediated effects on cell proliferation and migration in PDGF-BB-induced HA-VSMCs. MiR-1278 overexpression suppressed PDGF-BB-stimulated HA-VSMC proliferation and migration by targeting DNMT1. CONCLUSION CircPCNX promoted PDGF-BB-induced HA-VSMC proliferation and migration by elevating DNMT1 expression through sponging miR-1278.
Collapse
Affiliation(s)
- Wenbin Ma
- Department of Laboratory Medicine, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, Heilongjiang, China
| | - Dongmei Wei
- Department of Traditional Chinese Geriatric Medicine, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, Heilongjiang, China
| | - Xinying Li
- Department of Internal Medicine-Digestive, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, Heilongjiang, China
| | - Lina Shan
- Department of Internal Medicine-Cardiovascular, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, The Fourth Department of Internal Medicine-Cardiovascular Office, 10th Floor, Building 5, South Hospital of Qiqihar First Hospital, 700 Bukui South Street, Longsha District, Qiqihar, 161005, Heilongjiang, China
| | - Hua Fan
- Department of Laboratory Medicine, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, Heilongjiang, China
| | - Huixin Jin
- Department of Laboratory Medicine, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, 161005, Heilongjiang, China
| | - Binghui Song
- Department of Internal Medicine-Cardiovascular, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, The Fourth Department of Internal Medicine-Cardiovascular Office, 10th Floor, Building 5, South Hospital of Qiqihar First Hospital, 700 Bukui South Street, Longsha District, Qiqihar, 161005, Heilongjiang, China
| | - Ben Zhang
- Department of Internal Medicine-Cardiovascular, The First Hospital of Qiqihar/Affiliated Qiqihar Hospital, Southern Medical University, The Fourth Department of Internal Medicine-Cardiovascular Office, 10th Floor, Building 5, South Hospital of Qiqihar First Hospital, 700 Bukui South Street, Longsha District, Qiqihar, 161005, Heilongjiang, China.
| |
Collapse
|
109
|
Chen J, Yu H, Tan X, Mok SWF, Xie Y, Wang Y, Jiang X, Macrae VE, Lan L, Fu X, Zhu D. PFKFB3-driven vascular smooth muscle cell glycolysis promotes vascular calcification via the altered FoxO3 and lactate production. FASEB J 2023; 37:e23182. [PMID: 37682013 DOI: 10.1096/fj.202300900r] [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: 05/05/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023]
Abstract
A link between increased glycolysis and vascular calcification has recently been reported, but it remains unclear how increased glycolysis contributes to vascular calcification. We therefore investigated the role of PFKFB3, a critical enzyme of glycolysis, in vascular calcification. We found that PFKFB3 expression was upregulated in calcified mouse VSMCs and arteries. We showed that expression of miR-26a-5p and miR-26b-5p in calcified mouse arteries was significantly decreased, and a negative correlation between Pfkfb3 mRNA expression and miR-26a-5p or miR-26b-5p was seen in these samples. Overexpression of miR-26a/b-5p significantly inhibited PFKFB3 expression in VSMCs. Intriguingly, pharmacological inhibition of PFKFB3 using PFK15 or knockdown of PFKFB3 ameliorated vascular calcification in vD3 -overloaded mice in vivo or attenuated high phosphate (Pi)-induced VSMC calcification in vitro. Consistently, knockdown of PFKFB3 significantly reduced glycolysis and osteogenic transdifferentiation of VSMCs, whereas overexpression of PFKFB3 in VSMCs induced the opposite effects. RNA-seq analysis and subsequent experiments revealed that silencing of PFKFB3 inhibited FoxO3 expression in VSMCs. Silencing of FoxO3 phenocopied the effects of PFKFB3 depletion on Ocn and Opg expression but not Alpl in VSMCs. Pyruvate or lactate supplementation, the product of glycolysis, reversed the PFKFB3 depletion-mediated effects on ALP activity and OPG protein expression in VSMCs. Our results reveal that blockade of PFKFB3-mediated glycolysis inhibits vascular calcification in vitro and in vivo. Mechanistically, we show that FoxO3 and lactate production are involved in PFKFB3-driven osteogenic transdifferentiation of VSMCs. PFKFB3 may be a promising therapeutic target for the treatment of vascular calcification.
Collapse
Affiliation(s)
- Jiaxin Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Hongjiao Yu
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, China
| | - Xiao Tan
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Simon Wing Fai Mok
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Yuchen Xie
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yueheng Wang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xueyan Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Vicky E Macrae
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Lan Lan
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
110
|
Liu Q, Wang Y, Zhang T, Fang J, Meng S. Circular RNAs in vascular diseases. Front Cardiovasc Med 2023; 10:1247434. [PMID: 37840954 PMCID: PMC10570532 DOI: 10.3389/fcvm.2023.1247434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Vascular diseases are the leading cause of morbidity and mortality worldwide and are urgently in need of diagnostic biomarkers and therapeutic strategies. Circular RNAs (circRNAs) represent a unique class of RNAs characterized by a circular loop configuration and have recently been identified to possess a wide variety of biological functions. CircRNAs exhibit exceptional stability, tissue specificity, and are detectable in body fluids, thus holding promise as potential biomarkers. Their encoding function and stable gene expression also position circRNAs as an excellent alternative to gene therapy. Here, we briefly review the biogenesis, degradation, and functions of circRNAs. We summarize circRNAs discovered in major vascular diseases such as atherosclerosis and aneurysms, with a particular focus on molecular mechanisms of circRNAs identified in vascular endothelial cells and smooth muscle cells, in the hope to reveal new directions for mechanism, prognosis and therapeutic targets of vascular diseases.
Collapse
Affiliation(s)
| | | | | | | | - Shu Meng
- Department of Basic Science Research, Guangzhou Laboratory, Guangzhou, China
| |
Collapse
|
111
|
Zhu Y, Zhang JL, Yan XJ, Ji Y, Wang FF. Exploring a new mechanism between lactate and VSMC calcification: PARP1/POLG/UCP2 signaling pathway and imbalance of mitochondrial homeostasis. Cell Death Dis 2023; 14:598. [PMID: 37679327 PMCID: PMC10484939 DOI: 10.1038/s41419-023-06113-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
Lactate leads to the imbalance of mitochondria homeostasis, which then promotes vascular calcification. PARP1 can upregulate osteogenic genes and accelerate vascular calcification. However, the relationship among lactate, PARP1, and mitochondrial homeostasis is unclear. The present study aimed to explore the new molecular mechanism of lactate to promote VSMC calcification by evaluating PARP1 as a breakthrough molecule. A coculture model of VECs and VSMCs was established, and the model revealed that the glycolysis ability and lactate production of VECs were significantly enhanced after incubation in DOM. Osteogenic marker expression, calcium deposition, and apoptosis in VSMCs were decreased after lactate dehydrogenase A knockdown in VECs. Mechanistically, exogenous lactate increased the overall level of PARP and PARylation in VSMCs. PARP1 knockdown inhibited Drp1-mediated mitochondrial fission and partially restored PINK1/Parkin-mediated mitophagy, thereby reducing mitochondrial oxidative stress. Moreover, lactate induced the translocation of PARP1 from the nucleus to the mitochondria, which then combined with POLG and inhibited POLG-mediated mitochondrial DNA synthesis. This process led to the downregulation of mitochondria-encoded genes, disturbance of mitochondrial respiration, and inhibition of oxidative phosphorylation. The knockdown of PARP1 could partially reverse the damage of mitochondrial gene expression and function caused by lactate. Furthermore, UCP2 was upregulated by the PARP1/POLG signal, and UCP2 knockdown inhibited Drp1-mediated mitochondrial fission and partially recovered PINK1/Parkin-mediated mitophagy. Finally, UCP2 knockdown in VSMCs alleviated DOM-caused VSMC calcification in the coculture model. The study results thus suggest that upregulated PARP1 is involved in the mechanism through which lactate accelerates VSMC calcification partly via POLG/UCP2-caused unbalanced mitochondrial homeostasis.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Jia-Li Zhang
- Department of Gastroenterology Centre, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China
| | - Xue-Jiao Yan
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Yuan Ji
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China.
| | - Fang-Fang Wang
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China.
| |
Collapse
|
112
|
Rager C, Klöpper T, Pfeil U, Tasch S, Whittaker MR, Exintaris B, Mietens A, Middendorff R. Reference Gene U2 Enables Direct Comparison between Relative Gene Expression Levels of Vascular Smooth Muscle Cells in Tissue and Culture Using Real-Time Quantitative PCR. Cells 2023; 12:2135. [PMID: 37681867 PMCID: PMC10487071 DOI: 10.3390/cells12172135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
In nearly every lab, real-time quantitative polymerase chain reaction (qPCR) is used to quantify gene expression. However, a comparison of different samples requires the careful selection of suitable reference genes (RGs), sometimes referred to as housekeeping genes. In the case of vascular smooth muscle cells (vSMCs), it is important to know under which conditions gene expression in isolated and cultured vSMCs can be compared with vSMCs in a healthy blood vessel. We isolated the vSMC-containing layer of the rat aorta (tunica media) and used one (longitudinal) half for direct RNA extraction, while the other half served to isolate and culture vSMCs prior to RNA extraction. First, the expression of the routinely used RGs beta-actin (Actb) and Glyceraldehyde-3-phosphate dehydrogenase (Gapdh) is investigated in intact media and corresponding cultured vSMCs. Significant differences in their Ct values show that these RGs could not be used for such direct comparisons; therefore, we select 15 different RGs. Only the gene expression of the small ribonuclear protein (snRNP) U2 shows no significant differences between the absolute Ct values of cultured vSMCs and the intact media; moreover, no differences were found between male and female rats in our experimental setup. In conclusion, U2 was shown to be an appropriate (sex-independent) RG to compare relative expression levels of vSMCs in culture to those vSMCs within their physiological tissue environment.
Collapse
Affiliation(s)
- Christine Rager
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
- Drug Discovery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Melbourne, VIC 3052, Australia
| | - Tobias Klöpper
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| | - Uwe Pfeil
- Cardiopulmonary Neurobiology Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany
| | - Sabine Tasch
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| | - Michael Raymond Whittaker
- Drug Discovery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Melbourne, VIC 3052, Australia
| | - Betty Exintaris
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Melbourne, VIC 3052, Australia
| | - Andrea Mietens
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| | - Ralf Middendorff
- Signalling Transduction Group, Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany; (C.R.)
| |
Collapse
|
113
|
Koval OM, Nguyen EK, Mittauer DJ, Ait-Aissa K, Chinchankar WC, Grumbach IM. Regulation of Smooth Muscle Cell Proliferation by Mitochondrial Ca2+ in Type 2 Diabetes. Int J Mol Sci 2023; 24:12897. [PMID: 37629079 PMCID: PMC10454141 DOI: 10.3390/ijms241612897] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Type 2 diabetes (T2D) is associated with increased risk of atherosclerotic vascular disease due to excessive vascular smooth muscle cell (VSMC) proliferation. Here, we investigated the role of mitochondrial dysfunction and Ca2+ levels in VSMC proliferation in T2D. VSMCs were isolated from normoglycemic and T2D-like mice induced by diet. The effects of mitochondrial Ca2+ uptake were studied using mice with selectively inhibited mitochondrial Ca2+/calmodulin-dependent kinase II (mtCaMKII) in VSMCs. Mitochondrial transition pore (mPTP) was blocked using ER-000444793. VSMCs from T2D compared to normoglycemic mice exhibited increased proliferation and baseline cytosolic Ca2+ levels ([Ca2+]cyto). T2D cells displayed lower endoplasmic reticulum Ca2+ levels, reduced mitochondrial Ca2+ entry, and increased Ca2+ leakage through the mPTP. Mitochondrial and cytosolic Ca2+ transients were diminished in T2D cells upon platelet-derived growth factor (PDGF) administration. Inhibiting mitochondrial Ca2+ uptake or the mPTP reduced VSMC proliferation in T2D, but had contrasting effects on [Ca2+]cyto. In T2D VSMCs, enhanced activation of Erk1/2 and its upstream regulators was observed, driven by elevated [Ca2+]cyto. Inhibiting mtCaMKII worsened the Ca2+ imbalance by blocking mitochondrial Ca2+ entry, leading to further increases in [Ca2+]cyto and Erk1/2 hyperactivation. Under these conditions, PDGF had no effect on VSMC proliferation. Inhibiting Ca2+-dependent signaling in the cytosol reduced excessive Erk1/2 activation and VSMC proliferation. Our findings suggest that altered Ca2+ handling drives enhanced VSMC proliferation in T2D, with mitochondrial dysfunction contributing to this process.
Collapse
Affiliation(s)
- Olha M. Koval
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Emily K. Nguyen
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Dylan J. Mittauer
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Karima Ait-Aissa
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - William C. Chinchankar
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Isabella M. Grumbach
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
- Veterans Affairs Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
114
|
Yu Y, Wu XQ, Su FF, Yue CF, Zhou XM, Xu C. Maximakinin reduced intracellular Ca 2+ level in vascular smooth muscle cells through AMPK/ERK1/2 signaling pathways. Hypertens Res 2023; 46:1949-1960. [PMID: 37258626 DOI: 10.1038/s41440-023-01330-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 06/02/2023]
Abstract
We detect the antihypertensive effects of maximakinin (MK) on renal hypertensive rats (RHRs) and further research the influence of MK on vascular smooth muscle cells (VSMCs) to explore its hypotensive mechanism. The effects of MK on arterial blood pressure were observed in RHRs. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT) assays were performed to detect the effect of MK on VSMC viability. Western blot and flow cytometry were used to investigate the influence of MK on intracellular Ca2+ levels and protein expression changes in VSMCs. In addition, specific protein inhibitors were applied to confirm the involvement of Ca2+-related signaling pathways induced by MK in VSMCs. MK showed a more significant antihypertensive effect than bradykinin in RHRs. MK significantly decreased intracellular Ca2+ concentrations. Furthermore, MK significantly induced the phosphorylation of signaling molecules, including extracellular signal-regulated kinase 1/2 (ERK1/2), P38, AMP-activated protein kinase (AMPK) and Akt in VSMCs. Moreover, only ERK1/2 inhibitor U0126 and AMPK inhibitor Compound C completely restored the decreased intracellular Ca2+ level induced by MK, and further research demonstrated that AMPK functioned upstream of ERK1/2 following exposure to MK. Finally, HOE-140, an inhibitor of the bradykinin B2 receptors (B2Rs), was applied to investigate the potential targets of MK in VSMCs. HOE-140 significantly blocked the AMPK/ERK1/2 pathway induced by MK, suggesting that the B2Rs might play an important role in MK-induced AMPK and ERK1/2 activation. MK significantly reduces blood pressure in RHRs. MK exerts its antihypertensive effect by activating the B2Rs and downstream AMPK/ERK1/2 pathways, leading to significantly reduced Ca2+ levels in VSMCs.
Collapse
Affiliation(s)
- Yang Yu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Xue-Qian Wu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Fan-Fan Su
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Cai-Feng Yue
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Xiao-Mian Zhou
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China
| | - Cheng Xu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
115
|
Perry RN, Albarracin D, Aherrahrou R, Civelek M. Network Preservation Analysis Reveals Dysregulated Metabolic Pathways in Human Vascular Smooth Muscle Cell Phenotypic Switching. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:372-381. [PMID: 37387208 PMCID: PMC10434832 DOI: 10.1161/circgen.122.003781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/06/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Vascular smooth muscle cells are key players involved in atherosclerosis, the underlying cause of coronary artery disease. They can play either beneficial or detrimental roles in lesion pathogenesis, depending on the nature of their phenotypic changes. An in-depth characterization of their gene regulatory networks can help better understand how their dysfunction may impact disease progression. METHODS We conducted a gene expression network preservation analysis in aortic smooth muscle cells isolated from 151 multiethnic heart transplant donors cultured under quiescent or proliferative conditions. RESULTS We identified 86 groups of coexpressed genes (modules) across the 2 conditions and focused on the 18 modules that are least preserved between the phenotypic conditions. Three of these modules were significantly enriched for genes belonging to proliferation, migration, cell adhesion, and cell differentiation pathways, characteristic of phenotypically modulated proliferative vascular smooth muscle cells. The majority of the modules, however, were enriched for metabolic pathways consisting of both nitrogen-related and glycolysis-related processes. Therefore, we explored correlations between nitrogen metabolism-related genes and coronary artery disease-associated genes and found significant correlations, suggesting the involvement of the nitrogen metabolism pathway in coronary artery disease pathogenesis. We also created gene regulatory networks enriched for genes in glycolysis and predicted key regulatory genes driving glycolysis dysregulation. CONCLUSIONS Our work suggests that dysregulation of vascular smooth muscle cell metabolism participates in phenotypic transitioning, which may contribute to disease progression, and suggests that AMT (aminomethyltransferase) and MPI (mannose phosphate isomerase) may play an important role in regulating nitrogen and glycolysis-related metabolism in smooth muscle cells.
Collapse
Affiliation(s)
- R. Noah Perry
- Center for Public Health Genomics (R.N.P., R.A., M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (R.N.P., D.A., M.C.), University of Virginia, Charlottesville
| | - Diana Albarracin
- Department of Biomedical Engineering (R.N.P., D.A., M.C.), University of Virginia, Charlottesville
| | - Redouane Aherrahrou
- Center for Public Health Genomics (R.N.P., R.A., M.C.), University of Virginia, Charlottesville
| | - Mete Civelek
- Center for Public Health Genomics (R.N.P., R.A., M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (R.N.P., D.A., M.C.), University of Virginia, Charlottesville
| |
Collapse
|
116
|
Zhao Y, Zhao G, Chang Z, Zhu T, Zhao Y, Lu H, Xue C, Saunders TL, Guo Y, Chang L, Chen YE, Zhang J. Generating endogenous Myh11-driven Cre mice for sex-independent gene deletion in smooth muscle cells. JCI Insight 2023; 8:e171661. [PMID: 37289544 PMCID: PMC10443793 DOI: 10.1172/jci.insight.171661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
Specific and efficient smooth muscle cell-targeted (SMC-targeted) gene deletion is typically achieved by pairing SMMHC-CreERT2-Tg mice with mice carrying the loxP-flanked gene. However, the transgene, CreERT2, is not controlled by the endogenous Myh11 gene promoter, and the codon-modified iCreERT2 exhibits significant tamoxifen-independent leakage. Furthermore, because the Cre-bearing bacterial artificial chromosome (BAC) is inserted onto the Y chromosome, the SMMHC-CreERT2-Tg mice strain can only exhibit gene deletions in male mice. Additionally, there is a lack of Myh11-driven constitutive Cre mice when tamoxifen usage is a concern. We used CRISPR/Cas9-mediated homologous recombination between a donor vector carrying the CreNLSP2A or CreERT2-P2A sequence and homologous arm surrounding the translation start site of the Myh11 gene to generate Cre-knockin mice. The P2A sequence enables the simultaneous translation of Cre and endogenous proteins. Using reporter mice, we assessed Cre-mediated recombination efficiency, specificity, tamoxifen-dependent controllability, and functionality in both sexes. Both constitutive (Myh11-CreNLSP2A) and inducible (Myh11-CreERT2-P2A) Cre mice demonstrated efficient, SMC-specific, sex-independent Cre recombinase activity without confounding endogenous gene expression. Combined with recently generated BAC transgenic Myh11-CreERT2-RAD mice and the Itga8-CreERT2 mouse models, our models will help expand the research toolbox, facilitating unbiased and comprehensive research in SMCs and SMC-dependent cardiovascular diseases.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Pharmacology and
| | - Guizhen Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Ziyi Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Tianqing Zhu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Ying Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Chao Xue
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Thomas L. Saunders
- Transgenic Animal Model Core, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Lin Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
117
|
Li J, Li X, Song S, Sun Z, Li Y, Yang L, Xie Z, Cai Y, Zhao Y. Mitochondria spatially and temporally modulate VSMC phenotypes via interacting with cytoskeleton in cardiovascular diseases. Redox Biol 2023; 64:102778. [PMID: 37321061 DOI: 10.1016/j.redox.2023.102778] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023] Open
Abstract
Cardiovascular diseases caused by atherosclerosis (AS) seriously endanger human health, which is closely related to vascular smooth muscle cell (VSMC) phenotypes. VSMC phenotypic transformation is marked by the alteration of phenotypic marker expression and cellular behaviour. Intriguingly, the mitochondrial metabolism and dynamics altered during VSMC phenotypic transformation. Firstly, this review combs VSMC mitochondrial metabolism in three aspects: mitochondrial ROS generation, mutated mitochondrial DNA (mtDNA) and calcium metabolism respectively. Secondly, we summarized the role of mitochondrial dynamics in regulating VSMC phenotypes. We further emphasized the association between mitochondria and cytoskelton via presenting cytoskeletal support during mitochondrial dynamics process, and discussed its impact on their respective dynamics. Finally, considering that both mitochondria and cytoskeleton are mechano-sensitive organelles, we demonstrated their direct and indirect interaction under extracellular mechanical stimuli through several mechano-sensitive signaling pathways. We additionally discussed related researches in other cell types in order to inspire deeper thinking and reasonable speculation of potential regulatory mechanism in VSMC phenotypic transformation.
Collapse
Affiliation(s)
- Jingwen Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Xinyue Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Sijie Song
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhengwen Sun
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yuanzhu Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Long Yang
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhenhong Xie
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yikui Cai
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yinping Zhao
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
118
|
Fang Z, Wu G, Sheng J, Ye B, Huang Z, Xu J, Zhang J, Han J, Han B, Xu J. Gasdermin D affects aortic vascular smooth muscle cell pyroptosis and Ang II-induced vascular remodeling. Heliyon 2023; 9:e16619. [PMID: 37303505 PMCID: PMC10248119 DOI: 10.1016/j.heliyon.2023.e16619] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are primarily responsible for vasoconstriction and the regulation of blood pressure1. Pyroptosis, a particular form of regulated cell death, is involved in multiple vascular injuries, including hypertensive vascular dysfunction. This pyroptotic cell death is mediated by the pore-forming protein of Gasdermin D (GSDMD). This study was designed to examine the direct effect of GSDMD on smooth muscle cell pyroptosis and vascular remodeling. Findings revealed that GSDMD was activated in Angiotensin (Ang) II- treated aortas. We then showed that genetic deletion of Gsdmd reduced vascular remodeling and aorta pyroptosis induced by Ang II in vivo. Aberrant expression of GSDMD by recombinant AAV9 virus carrying Gsdmd cDNA aggravated the level of pyroptosis in aortas of Ang II mice. Gain- and loss-of- function analysis further confirmed that GSDMD regulated the pyroptosis of murine aortic vascular smooth muscle cells (MOVAS) in an in vitro model of tumor necrosis factor (TNF)-α treatment, which was achieved by transfecting expressing plasmid or siRNA, respectively. Overall, this study provided evidence supporting the active involvement of GSDMD in smooth muscle cell pyroptosis and Ang II-induced mice vascular injury. This finding lends credence to GSDMD as a potential therapeutic target for hypertensive vascular remodeling via inhibiting pyroptosis.
Collapse
Affiliation(s)
- Zimin Fang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Sheng
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouqing Huang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianjiang Xu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jianqin Zhang
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jibo Han
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Bingjiang Han
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jiajun Xu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
119
|
Wang Z, Ma J, Yue H, Zhang Z, Fang F, Wang G, Liu X, Shen Y. Vascular smooth muscle cells in intracranial aneurysms. Microvasc Res 2023:104554. [PMID: 37236346 DOI: 10.1016/j.mvr.2023.104554] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023]
Abstract
Intracranial aneurysm (IA) is a severe cerebrovascular disease characterized by abnormal bulging of cerebral vessels that may rupture and cause a stroke. The expansion of the aneurysm accompanies by the remodeling of vascular matrix. It is well-known that vascular remodeling is a process of synthesis and degradation of extracellular matrix (ECM), which is highly dependent on the phenotype of vascular smooth muscle cells (VSMCs). The phenotypic switching of VSMC is considered to be bidirectional, including the physiological contractile phenotype and alternative synthetic phenotype in response to injury. There is increasing evidence indicating that VSMCs have the ability to switch to various phenotypes, including pro-inflammatory, macrophagic, osteogenic, foamy and mesenchymal phenotypes. Although the mechanisms of VSMC phenotype switching are still being explored, it is becoming clear that phenotype switching of VSMCs plays an essential role in IA formation, progression, and rupture. This review summarized the various phenotypes and functions of VSMCs associated with IA pathology. The possible influencing factors and potential molecular mechanisms of the VSMC phenotype switching were further discussed. Understanding how phenotype switching of VSMC contributed to the pathogenesis of unruptured IAs can bring new preventative and therapeutic strategies for IA.
Collapse
Affiliation(s)
- Zhenye Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Jia Ma
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Hongyan Yue
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Zhewei Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Guixue Wang
- Jinfeng Laboratory, Chongqing 401329, China; Key Laboratory of Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
120
|
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
|
121
|
Derhambakhsh S, Mohammadi J, Shokrgozar MA, Rabbani H, Sadeghi N, Nekounam H, Mohammadi S, Lee KB, Khakbiz M. Investigation of electrical stimulation on phenotypic vascular smooth muscle cells differentiation in tissue-engineered small-diameter vascular graft. Tissue Cell 2023; 81:101996. [PMID: 36657256 DOI: 10.1016/j.tice.2022.101996] [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: 07/18/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
In the development of vascular tissue engineering, particularly in the case of small diameter vessels, one of the key obstacles is the blockage of these veins once they enter the in vivo environment. One of the contributing factors to this problem is the aberrant proliferation and migration of vascular smooth muscle cells (VSMCs) from the media layer of the artery to the interior of the channel. Two distinct phenotypes have been identified for smooth muscle cells, namely synthetic and contractile. Since the synthetic phenotype plays an essential role in the unusual growth and migration, the aim of this study was to convert the synthetic phenotype into the contractile one, which is a solution to prevent the abnormal growth of VSMCs. To achieve this goal, these cells were subjected to electrical signals, using a 1000 μA sinusoidal stimulation at 10 Hz for four days, with 20 min duration per 24 h. The morphological transformations and changes in the expression of vimentin, nestin, and β-actin proteins were then studied using ICC and flow cytometry assays. Also, the expression of VSMC specific markers such as smooth muscle myosin heavy chain (SMMHC) and smooth muscle alpha-actin (α-SMA) were evaluated using RT-PCR test. In the final phase of this study, the sheep decellularized vessel was employed as a scaffold for seeding these cells. Based on the results, electrical stimulation resulted in some morphological alterations in VSMCs. Furthermore, the observed reductions in the expression levels of vimentin, nestin and β-actin proteins and increase in the expression of SMMHC and α-SMA markers showed that it is possible to convert the synthetic phenotype to the contractile one using the studied regime of electrical stimulation. Finally, it can be concluded that electrical stimulation can significantly affect the phenotype of VSMCs, as demonstrated in this study.
Collapse
Affiliation(s)
- Sara Derhambakhsh
- Division of Biomedical Engineering, Department of Life Science, Faculty of New Sciences and Technologies, University of Tehran, Tehran 439957131, Iran
| | - Javad Mohammadi
- Division of Biomedical Engineering, Department of Life Science, Faculty of New Sciences and Technologies, University of Tehran, Tehran 439957131, Iran.
| | | | - Hodjattallah Rabbani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Niloufar Sadeghi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Houra Nekounam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Sotoudeh Mohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Mehrdad Khakbiz
- Division of Biomedical Engineering, Department of Life Science, Faculty of New Sciences and Technologies, University of Tehran, Tehran 439957131, Iran.
| |
Collapse
|
122
|
Anderson JR, Morin EE, Brayer KJ, Salbato S, Gonzalez Bosc LV, Kanagy NL, Naik JS. Single-cell transcriptomic heterogeneity between conduit and resistance mesenteric arteries in rats. Physiol Genomics 2023; 55:179-193. [PMID: 36912534 PMCID: PMC10085562 DOI: 10.1152/physiolgenomics.00126.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
The endothelium contains morphologically similar cells throughout the vasculature, but individual cells along the length of a single vascular tree or in different regional circulations function dissimilarly. When observations made in large arteries are extrapolated to explain the function of endothelial cells (ECs) in the resistance vasculature, only a fraction of these observations are consistent between artery sizes. To what extent endothelial (EC) and vascular smooth muscle cells (VSMCs) from different arteriolar segments of the same tissue differ phenotypically at the single-cell level remains unknown. Therefore, single-cell RNA-seq (10x Genomics) was performed using a 10X Genomics Chromium system. Cells were enzymatically digested from large (>300 µm) and small (<150 µm) mesenteric arteries from nine adult male Sprague-Dawley rats, pooled to create six samples (3 rats/sample, 3 samples/group). After normalized integration, the dataset was scaled before unsupervised cell clustering and cluster visualization using UMAP plots. Differential gene expression analysis allowed us to infer the biological identity of different clusters. Our analysis revealed 630 and 641 differentially expressed genes (DEGs) between conduit and resistance arteries for ECs and VSMCs, respectively. Gene ontology analysis (GO-Biological Processes, GOBP) of scRNA-seq data discovered 562 and 270 pathways for ECs and VSMCs, respectively, that differed between large and small arteries. We identified eight and seven unique ECs and VSMCs subpopulations, respectively, with DEGs and pathways identified for each cluster. These results and this dataset allow the discovery and support of novel hypotheses needed to identify mechanisms that determine the phenotypic heterogeneity between conduit and resistance arteries.
Collapse
Affiliation(s)
- Jacob R Anderson
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Emily E Morin
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Kathryn J Brayer
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Sophia Salbato
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Laura V Gonzalez Bosc
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Jay S Naik
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| |
Collapse
|
123
|
Non-genomic Effect of Estradiol on the Neurovascular Unit and Possible Involvement in the Cerebral Vascular Accident. Mol Neurobiol 2023; 60:1964-1985. [PMID: 36596967 DOI: 10.1007/s12035-022-03178-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
Cerebrovascular diseases, such as ischemic cerebral vascular accident (CVA), are responsible for causing high rates of morbidity, mortality, and disability in the population. The neurovascular unit (NVU) during and after ischemic CVA plays crucial roles in cell regulation and preservation, the immune and inflammatory response, and cell and/or tissue survival and repair. Cellular responses to 17β-estradiol (E2) can be triggered by two mechanisms: one called classical or genomic, which is due to the activation of the "classical" nuclear estrogen receptors α (ERα) and β (ERβ), and the non-genomic or rapid mechanism, which is due to the activation of the G protein-coupled estrogen receptor 1 (GPER) that is located in the plasma membrane and some in intracellular membranes, such as in the Golgi apparatus and endoplasmic reticulum. Nuclear receptors can regulate gene expression and cellular functions. On the contrary, activating the GPER by E2 and/or its G-1 agonist triggers several rapid cell signaling pathways. Therefore, E2 or its G-1 agonist, by mediating GPER activation and/or expression, can influence several NVU cell types. Most studies argue that the activation of the GPER may be used as a potential therapeutic target in various pathologies, such as CVA. Thus, with this review, we aimed to summarize the existing literature on the role of GPER mediated by E2 and/or its agonist G-1 in the physiology and pathophysiology of NVU.
Collapse
|
124
|
Li L, Mou J, Han Y, Wang M, Lu S, Ma Q, Wang J, Ye J, Sun G. Calenduloside e modulates macrophage polarization via KLF2-regulated glycolysis, contributing to attenuates atherosclerosis. Int Immunopharmacol 2023; 117:109730. [PMID: 36878047 DOI: 10.1016/j.intimp.2023.109730] [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: 10/03/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 03/06/2023]
Abstract
Glycolysis-mediated macrophage polarization plays a crucial role in atherosclerosis. Although it is known that calenduloside E (CE) exerts anti-inflammatory and lipid-lowering effects in atherosclerosis, the underlying mechanism of action is not clearly understood. We hypothesized that CE functions by inhibiting M1 macrophage polarization via regulation of glycolysis. To verify this hypothesis, we determined the effects of CE in apolipoprotein E deficient (ApoE-/-) mice and on macrophage polarization in oxidized low-density lipoprotein (ox-LDL)-induced RAW 264.7 macrophages and peritoneal macrophages. We also determined whether these effects are linked to regulation of glycolysis both in vivo and in vitro. The plaque size was reduced, and serum cytokine levels were decreased in the ApoE-/- +CE group compared with that in the model group. CE decreased lipid droplet formation, inflammatory factor levels, and mRNA levels of M1 macrophage markers in ox-ldl-induced macrophages. CE suppressed ox-ldl-induced glycolysis, lactate levels, and glucose uptake. The relationship between glycolysis and M1 macrophage polarization was demonstrated using the glycolysis inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one. CE substantially upregulated ox-ldl-induced Kruppel-like transcription factor (KLF2) expression, and the effects of CE on ox-ldl-induced glycolysis and inflammatory factor levels disappeared after KLF2 knockdown. Together, our findings suggest that CE alleviates atherosclerosis by inhibiting glycolysis-mediated M1 macrophage polarization through upregulation of KLF2 expression, providing a new strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Lanfang Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Junyu Mou
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Yanwei Han
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Min Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shan Lu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Qiuxiao Ma
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jialu Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingxue Ye
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
125
|
Mengozzi A, Costantino S, Mongelli A, Mohammed SA, Gorica E, Delfine V, Masi S, Virdis A, Ruschitzka F, Paneni F. Epigenetic Signatures in Arterial Hypertension: Focus on the Microvasculature. Int J Mol Sci 2023; 24:ijms24054854. [PMID: 36902291 PMCID: PMC10003673 DOI: 10.3390/ijms24054854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Systemic arterial hypertension (AH) is a multifaceted disease characterized by accelerated vascular aging and high cardiometabolic morbidity and mortality. Despite extensive work in the field, the pathogenesis of AH is still incompletely understood, and its treatment remains challenging. Recent evidence has shown a deep involvement of epigenetic signals in the regulation of transcriptional programs underpinning maladaptive vascular remodeling, sympathetic activation and cardiometabolic alterations, all factors predisposing to AH. After occurring, these epigenetic changes have a long-lasting effect on gene dysregulation and do not seem to be reversible upon intensive treatment or the control of cardiovascular risk factors. Among the factors involved in arterial hypertension, microvascular dysfunction plays a central role. This review will focus on the emerging role of epigenetic changes in hypertensive-related microvascular disease, including the different cell types and tissues (endothelial cells, vascular smooth muscle cells and perivascular adipose tissue) as well as the involvement of mechanical/hemodynamic factors, namely, shear stress.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Shafeeq A. Mohammed
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, 8091 Zurich, Switzerland
- Correspondence: or francesco.paneni@uzh; Tel.: +41-44-6355096
| |
Collapse
|
126
|
Koval OM, Nguyen EK, Mittauer DJ, Ait-Aissa K, Chinchankar W, Qian L, Madesh M, Dai DF, Grumbach IM. The mitochondrial regulation of smooth muscle cell proliferation in type 2 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528765. [PMID: 36824758 PMCID: PMC9948984 DOI: 10.1101/2023.02.15.528765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Background Type 2 diabetes (T2D) is associated with a strongly increased risk for restenosis after angioplasty driven by proliferation of vascular smooth muscle cells (VSMCs). Here, we sought to determine whether and how mitochondrial dysfunction in T2D drives VSMC proliferation with a focus on ROS and intracellular [Ca 2+ ] that both drive cell proliferation, occur in T2D and are regulated by mitochondrial activity. Methods Using a diet-induced mouse model of T2D, the inhibition of the mitochondrial Ca 2+ /calmodulin-dependent kinase II (mtCaMKII), a regulator of Ca 2+ entry via the mitochondrial Ca 2+ uniporter selectively in VSMCs, we performed in vivo phenotyping after mechanical injury and established the mechanisms of excessive proliferation in cultured VSMCs. Results In T2D, the inhibition of mtCaMKII reduced both neointima formation after mechanical injury and the proliferation of cultured VSMCs. VSMCs from T2D mice displayed accelerated proliferation, reduced mitochondrial Ca 2+ entry and membrane potential with elevated baseline [Ca 2+ ] cyto compared to cells from normoglycemic mice. Accelerated proliferation after PDGF treatment was driven by activation of Erk1/2 and its upstream regulators. Hyperactivation of Erk1/2 was Ca 2+ -dependent rather than mitochondrial ROS-driven Ca 2+ -dependent and included the activation of CaMKII in the cytosol. The inhibition of mtCaMKII exaggerated the Ca 2+ imbalance by lowering mitochondrial Ca 2+ entry and increasing baseline [Ca 2+ ] cyto , further enhancing baseline Erk1/2 activation. With inhibition of mtCaMKII, PDGF treatment had no additional effect on cell proliferation. Inhibition of activated CaMKII in the cytosol decreased excessive Erk1/2 activation and reduced VSMC proliferation. Conclusions Collectively, our results provide evidence for the molecular mechanisms of enhanced VSMC proliferation after mechanical injury by mitochondrial Ca 2+ entry in T2D.
Collapse
Affiliation(s)
- Olha M. Koval
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Emily K. Nguyen
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Dylan J. Mittauer
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Karima Ait-Aissa
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - William Chinchankar
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Lan Qian
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Muniswamy Madesh
- Center for Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229, USA
| | - Dao-Fu Dai
- Division of Pathology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
| | - Isabella M. Grumbach
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City IA 52242, USA
- Veterans Affairs Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
127
|
Zhu T, Hu Q, Yuan Y, Yao H, Zhang J, Qi J. Mitochondrial dynamics in vascular remodeling and target-organ damage. Front Cardiovasc Med 2023; 10:1067732. [PMID: 36860274 PMCID: PMC9970102 DOI: 10.3389/fcvm.2023.1067732] [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: 10/12/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Vascular remodeling is the pathological basis for the development of many cardiovascular diseases. The mechanisms underlying endothelial cell dysfunction, smooth muscle cell phenotypic switching, fibroblast activation, and inflammatory macrophage differentiation during vascular remodeling remain elusive. Mitochondria are highly dynamic organelles. Recent studies showed that mitochondrial fusion and fission play crucial roles in vascular remodeling and that the delicate balance of fusion-fission may be more important than individual processes. In addition, vascular remodeling may also lead to target-organ damage by interfering with the blood supply to major body organs such as the heart, brain, and kidney. The protective effect of mitochondrial dynamics modulators on target-organs has been demonstrated in numerous studies, but whether they can be used for the treatment of related cardiovascular diseases needs to be verified in future clinical studies. Herein, we summarize recent advances regarding mitochondrial dynamics in multiple cells involved in vascular remodeling and associated target-organ damage.
Collapse
Affiliation(s)
- Tong Zhu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingxun Hu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai, China,Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Yanggang Yuan
- Department of Nephrology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Huijuan Yao
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhang
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Jian Zhang,
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Jia Qi,
| |
Collapse
|
128
|
Liu H, Yin H, Wang Z, Yuan Q, Xu F, Chen Y, Li C. Rho A/ROCK1 signaling-mediated metabolic reprogramming of valvular interstitial cells toward Warburg effect accelerates aortic valve calcification via AMPK/RUNX2 axis. Cell Death Dis 2023; 14:108. [PMID: 36774349 PMCID: PMC9922265 DOI: 10.1038/s41419-023-05642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/13/2023]
Abstract
The aberrant differentiation of valvular interstitial cells (VICs) to osteogenic lineages promotes calcified aortic valves disease (CAVD), partly activated by potentially destructive hemodynamic forces. These involve Rho A/ROCK1 signaling, a mechano-sensing pathway. However, how Rho A/ROCK1 signaling transduces mechanical signals into cellular responses and disrupts normal VIC homeostasis remain unclear. We examined Rho A/ROCK1 signaling in human aortic valves, and further detected how Rho A/ROCK1 signaling regulates mineralization in human VICs. Aortic valves (CAVD n = 22, normal control (NC) n = 12) from patients undergoing valve replacement were investigated. Immunostaining and western blotting analysis indicated that Rho A/ROCK1 signaling, as well as key transporters and enzymes involved in the Warburg effect, were markedly upregulated in human calcified aortic valves compared with those in the controls. In vitro, Rho A/ROCK1-induced calcification was confirmed as AMPK-dependent, via a mechanism involving metabolic reprogramming of human VICs to Warburg effect. Y-27632, a selective ROCK1 inhibitor, suppressed the Warburg effect, rescued AMPK activity and subsequently increased RUNX2 ubiquitin-proteasome degradation, leading to decreased RUNX2 protein accumulation in human VICs under pathological osteogenic stimulus. Rho A/ROCK1 signaling, which is elevated in human calcified aortic valves, plays a positive role in valvular calcification, partially through its ability to drive metabolic switching of VICs to the Warburg effect, leading to altered AMPK activity and RUNX2 protein accumulation. Thus, Rho A/ROCK1 signaling could be an important and unrecognized hub of destructive hemodynamics and cellular aerobic glycolysis that is essential to promote the CAVD process.
Collapse
Affiliation(s)
- Huiruo Liu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hang Yin
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhen Wang
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiuhuan Yuan
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Feng Xu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuguo Chen
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Chuanbao Li
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
129
|
Talamanca L, Gobet C, Naef F. Sex-dimorphic and age-dependent organization of 24-hour gene expression rhythms in humans. Science 2023; 379:478-483. [PMID: 36730411 DOI: 10.1126/science.add0846] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The circadian clock modulates human physiology. However, the organization of tissue-specific gene expression rhythms and how these depend on age and sex is not defined in humans. We combined data from the Genotype-Tissue Expression (GTEx) project with an algorithm that assigns circadian phases to 914 donors, by integrating temporal information from multiple tissues in each individual, to identify messenger RNA (mRNA) rhythms in 46 tissues. Clock transcripts showed conserved timing relationships and tight synchrony across the body. mRNA rhythms varied in breadth, covering global and tissue-specific functions, including metabolic pathways and systemic responses. The clock structure was conserved across sexes and age groups. However, overall gene expression rhythms were highly sex-dimorphic and more sustained in females. Rhythmic programs generally dampened with age across the body.
Collapse
Affiliation(s)
- Lorenzo Talamanca
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Cédric Gobet
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
130
|
Tan JL, Yi J, Cao XY, Wang FY, Xie SL, Zhou LL, Qin L, Dai AG. Celastrol: The new dawn in the treatment of vascular remodeling diseases. Biomed Pharmacother 2023; 158:114177. [PMID: 36809293 DOI: 10.1016/j.biopha.2022.114177] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Evidence is mounting that abnormal vascular remodeling leads to many cardiovascular diseases (CVDs). This suggests that vascular remodeling can be a crucial target for the prevention and treatment of CVDs. Recently, celastrol, an active ingredient of the broadly used Chinese herb Tripterygium wilfordii Hook F, has attracted extensive interest for its proven potential to improve vascular remodeling. Substantial evidence has shown that celastrol improves vascular remodeling by ameliorating inflammation, hyperproliferation, and migration of vascular smooth muscle cells, vascular calcification, endothelial dysfunction, extracellular matrix remodeling, and angiogenesis. Moreover, numerous reports have proven the positive effects of celastrol and its therapeutic promise in treating vascular remodeling diseases such as hypertension, atherosclerosis, and pulmonary artery hypertension. The present review summarizes and discusses the molecular mechanism of celastrol regulating vascular remodeling and provides preclinical proof for future clinical applications of celastrol.
Collapse
Affiliation(s)
- Jun-Lan Tan
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Jian Yi
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Xian-Ya Cao
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Fei-Ying Wang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Si-Lin Xie
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Ling-Ling Zhou
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Li Qin
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Ai-Guo Dai
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China.
| |
Collapse
|
131
|
Jiang F, Zhang B, Zhang X, Zhang R, Lu Q, Shi F, Xu J, Deng L. miRNA‑92a inhibits vascular smooth muscle cell phenotypic modulation and may help prevent in‑stent restenosis. Mol Med Rep 2023; 27:40. [PMID: 36601739 PMCID: PMC9835053 DOI: 10.3892/mmr.2023.12927] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/23/2022] [Indexed: 01/03/2023] Open
Abstract
The modulation of vascular smooth muscle cell (VSMC) phenotype during cellular proliferation and migration may represent a potential therapeutic approach for vascular intimal hyperplasia prevention. However, the precise role of this process in VSMC biology and remodeling remains unclear. In the present study, western blotting, PCR, MTT and Transwell assays were used to analyze related protein and mRNA expression, cell viability and cell migration, respectively. It was demonstrated that miR‑92a modulated VSMCs into a synthetic phenotype via the Kruppel‑like factor 4 (KLF4) pathway. Targeting microRNA (miRNA/miR)‑92a in VSMCs using a KLF4 inhibitor suppressed the synthetic phenotype and inhibited VSMC proliferation and migration. To further confirm this finding, the expression levels of miR‑92a were measured in patients undergoing coronary artery intervention. The serum miR‑92a expression levels were significantly higher in patients with in‑stent restenosis (ISR) compared with those in patients without ISR, whereas KLF4 expression was significantly reduced in the non‑ISR group. Bioinformatic analysis and promoter‑luciferase reporter assays were used to examine the regulatory mechanisms underlying KLF4 expression. KLF4 was demonstrated to be transcriptionally upregulated by miR‑92a in VSMCs. miRNA transfection was also performed to regulate the level of miR‑92a expression. miR‑92a overexpression inhibited VSMC proliferation and migration, and also increased the mRNA and protein expression levels of certain differentiated VSMC‑related genes. Finally, miR‑92a inhibition promoted the proliferation and migration of VSMCs, which could be reversed using a KLF4 inhibitor. Collectively, these results indicated that the local delivery of a KLF4 inhibitor may act as a novel therapeutic option for the prevention of ISR.
Collapse
Affiliation(s)
- Fenfen Jiang
- Department of Cardiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China,Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Bin Zhang
- Department of Anaesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiangyu Zhang
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Ran Zhang
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Qin Lu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Fengjie Shi
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Jianjiang Xu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Lang Deng
- Department of Cardiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313003, P.R. China,Correspondence to: Dr Lang Deng, Department of Cardiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, 198 Hongqi Road, Huzhou, Zhejiang 313003, P.R. China, E-mail:
| |
Collapse
|
132
|
Chen Y, Han Y, Wu Y, Hui R, Yang Y, Zhong Y, Zhang S, Zhang W. Pharmacogenetic association of the NR1H3 promoter variant with antihypertensive response among patients with hypertension: A longitudinal study. Front Pharmacol 2023; 14:1083134. [PMID: 36950018 PMCID: PMC10025344 DOI: 10.3389/fphar.2023.1083134] [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: 10/28/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Background: The genetic factors in assessing therapeutic efficacy and predicting antihypertensive drug response are unclear. Therefore, this study aims to identify the associations between variants and antihypertensive drug response. Methods: A longitudinal study including 1837 hypertensive patients was conducted in Northern China and followed up for a median 2.24 years. The associations of 11 candidate variants with blood pressure changes in response to antihypertensive drugs and with the risk of cardiovascular events during the follow-up were examined. The dual-luciferase assay was carried out to assess the effect of genetic variants on gene transcriptional activity. Results: The variant rs11039149A>G in the promoter of nuclear receptor subfamily 1 group H member 3 (NR1H3) was associated with the change in systolic blood pressure (ΔSBP) in response to calcium channel blockers (CCBs) monotherapy. Patients carrying rs11039149AG genotype showed a significant increase of systolic blood pressure (SBP) at follow-up compared with AA carriers, and the difference of ΔSBP between AG and AA carriers was 5.94 mm Hg (95%CI: 2.09-9.78, p = 0.002). In 1,184 patients with CCBs therapy, SBP levels decreased in AA carriers, but increased in AG carriers, the difference of ΔSBP between AG and AA carriers was 8.04 mm Hg (95%CI: 3.28-12.81, p = 0.001). Further analysis in 359 patients with CCBs monotherapy, the difference of ΔSBP between AG and AA carriers was 15.25 mm Hg (95%CI: 6.48-24.02, p = 0.001). However, there was no significant difference in ΔSBP between AG and AA carriers with CCBs multitherapy. The rs11039149A>G was not associated with the cardiovascular events incidence during the follow-up. Additionally, transcriptional factor forkhead box C1 (FOXC1) bound to the NR1H3 promoter containing rs11039149A and significantly increased the transcriptional activity, while rs11039149 A to G change led to a loss-of-function and disabled FOXC1 binding. For the other 10 variants, associations with blood pressure changes or risk of cardiovascular events were not observed. Conclusion: Hypertensive patients with rs11039149AG genotype in the NR1H3 gene have a significant worse SBP control in response to CCBs monotherapy compared with AA carriers. Our findings suggest that the NR1H3 gene might act as a promising genetic factor to affect individual sensitivity to antihypertensive drugs.
Collapse
Affiliation(s)
- Yu Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yuqing Han
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yiyi Wu
- The First Affiliated Hospital of Anhui University of Science and Technology, The First People’s Hospital of Huainan City, Huainan, Anhui, China
| | - Rutai Hui
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yunyun Yang
- Clinical Laboratory, Xiamen Key Laboratory of Genetic Testing, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yixuan Zhong
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Shuyuan Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Weili Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
- Central-China Branch of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Hospital, Zhengzhou, China
- *Correspondence: Weili Zhang,
| |
Collapse
|
133
|
Li G, Chen Q. lncRNA PCA3 Suppressed Carotid Artery Stenosis and Vascular Smooth Muscle Cell Function via Negatively Modulating the miR-124-3p/ITGB1 Axis. Clin Appl Thromb Hemost 2023; 29:10760296231190383. [PMID: 37583257 PMCID: PMC10467385 DOI: 10.1177/10760296231190383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/14/2023] [Accepted: 07/10/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND & OBJECTIVES Due to the hidden pathogen, carotid artery stenosis (CAS) always occurred at an advanced stage leading to serious sequelae and even deaths. The significance of long noncoding RNA (lncRNA) prostate cancer antigen 3 (PCA3) in CAS incidence and progression were evaluated aiming to explore a potential target for its therapy. MATERIALS AND METHODS Serum samples were collected from 83 asymptomatic CAS patients and 52 healthy individuals and PCA3 was compared using polymerase chain reaction (PCR). The PCA3 levels were compared between stable and unstable plaque in CAS patients. The effect of PCA3 on vascular smooth muscle cells (VSMCs) proliferation and motility was assessed by CCK8 and transwell assay. RESULTS PCA3 was downregulated in CAS patients and their unstable plaque tissues compared with healthy individuals and stable plaque, respectively. Reduced PCA3 could discriminate CAS patients with relatively high sensitivity and specificity and were associated with higher total cholesterol level and stenosis degree, unstable plaque, and complications. PCA3 downregulation predicted the adverse outcomes of CAS patients. In VSMCs, overexpressing PCA3 significantly suppressed cell proliferation, migration, and invasion, which was alleviated by miR-124-3p/ITGB1 axis. CONCLUSION PCA3 served as a biomarker of CAS and regulates the function of VSMCs through sponging miR-124-3p/ITGB1 and indirectly influence the stability of plaque.
Collapse
Affiliation(s)
- Guosheng Li
- The Affiliated Taizhou Second People's Hospital of Yangzhou University, Taizhou, China
| | - Qiang Chen
- Department of Neurology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| |
Collapse
|
134
|
Martín-González C, Godoy-Reyes AM, Abreu-González P, Fernández-Rodríguez CM, Martín-Ponce E, Sánchez-Pérez MJ, Alvisa-Negrín JC, Rodríguez-Gaspar M, González-Reimers E. Sclerostin, vascular risk factors, and brain atrophy in excessive drinkers. Front Hum Neurosci 2023; 17:1084756. [PMID: 36895513 PMCID: PMC9989031 DOI: 10.3389/fnhum.2023.1084756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/31/2023] [Indexed: 02/23/2023] Open
Abstract
Objective Heavy alcohol consumption causes several organic complications, including vessel wall calcification. Vascular damage may be involved in the development of brain atrophy and cognitive impairment. Recently, sclerostin (whose levels may be altered in alcoholics) has emerged as a major vascular risk factor. The objective of the present study is to analyze the prevalence of vascular calcifications in alcoholics, and the relationships of these lesions with brain atrophy, as well as the role of sclerostin on these alterations. Patients and methods A total of 299 heavy drinkers and 32 controls were included. Patients underwent cranial computed tomography, and several indices related to brain atrophy were calculated. In addition, patients and controls underwent plain radiography and were evaluated for the presence or absence of vascular calcium deposits, cardiovascular risk factors, liver function, alcohol intake, serum sclerostin, and routine laboratory variables. Results A total of 145 (48.47%) patients showed vascular calcium deposits, a proportion significantly higher than that observed in controls (χ2 = 16.31; p < 0.001). Vascular calcium deposits were associated with age (t = 6.57; p < 0.001), hypertension (t = 5.49; p < 0.001), daily ethanol ingestion (Z = 2.18; p = 0.029), duration of alcohol consumption (Z = 3.03; p = 0.002), obesity (χ2 = 4.65; p = 0.031), total cholesterol (Z = 2.04; p = 0.041), triglycerides (Z = 2.05; p = 0.04), and sclerostin levels (Z = 2.64; p = 0.008). Calcium deposits were significantly related to Bifrontal index (Z = 2.20; p = 0.028) and Evans index (Z = 2.25; p = 0.025). Serum sclerostin levels were related to subcortical brain atrophy, assessed by cella media index (Z = 2.43; p = 0.015) and Huckmann index (ρ = 0.204; p = 0.024). Logistic regression analyses disclosed that sclerostin was the only variable independently related to brain atrophy assessed by altered cella media index. Sclerostin was also related to the presence of vascular calcifications, although this relationship was displaced by age if this variable was also included. Conclusion Prevalence of vascular calcification in alcoholics is very high. Vascular calcium deposits are related to brain atrophy. Serum sclerostin is strongly related to brain shrinkage and also shows a significant relationship with vascular calcifications, only displaced by advanced age.
Collapse
Affiliation(s)
- Candelaria Martín-González
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - Ana María Godoy-Reyes
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - Pedro Abreu-González
- Departamento de Ciencias Médicas Básicas, Unidad de Fisiología, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Camino María Fernández-Rodríguez
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - Esther Martín-Ponce
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - María José Sánchez-Pérez
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - Julio César Alvisa-Negrín
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - Melchor Rodríguez-Gaspar
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| | - Emilio González-Reimers
- Departamento de Medicina Interna, Universidad de La Laguna, Servicio de Medicina Interna, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Spain
| |
Collapse
|
135
|
Xiong Q, Lu F, Xie X, Zhou W. Hypoxia-induced endothelial cell-derived exosome stimulates vascular smooth muscle cell proliferation and migration. Biomed Res 2023; 44:245-255. [PMID: 38008423 DOI: 10.2220/biomedres.44.245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
This study mainly used human VSMCs and ECs cultured in vitro to investigate whether exosomes (Exos) are involved in the communication between ECs and VSMCs under hypoxia, and to explore the role and mechanism of ECs-derived exosomes in the abnormal proliferation of VSMCs. VSMCs proliferation and migration were assessed by a series of cell function assays after culturing VSMCs alone or co-culturing ECs under hypoxia or normoxia. Next, exosomes were extracted from ECs under hypoxia or normoxia and characterized. We then introduced ECs-Exos to observe their effects on VSMCs proliferation and migration, and further evaluated the expression of transforming growth factor-beta receptor 1 (TGFBR1) pathway-related proteins. Finally, the effect of ECs-Exos on VSMCs function was evaluated after knocking down TGFBR1 in ECs. VSMCs treated with ECs-Exos exhibited increased proliferation and migration ability in hypoxic environment, and the expression of TGFBR1 pathway-related proteins was upregulated. Administration of ECs-Exos with TGFβ1 knockdown conspicuously reversed the promoting effects of ECs-Exos on cell proliferation and migration under hypoxia. In summary, hypoxia affected the secretion of extracellular vesicles by endothelial cells, which can be internalized by VSMCs and accelerate the abnormal proliferation and migration of VSMCs by delivering TGFBR1.
Collapse
Affiliation(s)
- Qinggen Xiong
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| | - Fei Lu
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| | - Xiaoming Xie
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| | - Wei Zhou
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| |
Collapse
|
136
|
Zou X, Liao Y, Liu Z, Xu X, Sun W, Qin H, Wang H, Liu J, Jing T. Exosomes Derived from AT2R-Overexpressing BMSC Prevent Restenosis After Carotid Artery Injury by Attenuating the Injury-Induced Neointimal Hyperplasia. J Cardiovasc Transl Res 2023; 16:112-126. [PMID: 35900670 PMCID: PMC9944384 DOI: 10.1007/s12265-022-10293-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022]
Abstract
Restenosis is a severe complication after percutaneous transluminal coronary angioplasty which limits the long-term efficacy of the intervention. In this study, we investigated the efficiency of exosomes derived from AT2R-overexpressing bone mesenchymal stem cells on the prevention of restenosis after carotid artery injury. Our data showed that AT2R-EXO promoted the proliferation and migration of vascular endothelial cells and maintained the ratio of eNOS/iNOS. On the contrary, AT2R-EXO inhibited the proliferation and migration of vascular smooth muscle cells. In vivo study proved that AT2R-Exo were more effectively accumulated in the injured carotid artery than EXO and Vehicle-EXO controls. AT2R-EXO treatment could improve blood flow of the injured carotid artery site more effectively. Further analysis revealed that AT2REXO prevents restenosis after carotid artery injury by attenuating the injury-induced neointimal hyperplasia. Our study provides a novel and more efficient exosome for the treatment of restenosis diseases after intervention.
Collapse
Affiliation(s)
- Xinliang Zou
- Department of Cardiology, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Yi Liao
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Zhihui Liu
- Department of Cardiology, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Xiang Xu
- Department of Cardiology, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Weiwei Sun
- Department of Cardiology, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Haoran Qin
- Department of Cardiology, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Haidong Wang
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Jianping Liu
- Department of Cardiology, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Tao Jing
- Department of Cardiology, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
137
|
Differential Regulation of MMPs, Apoptosis and Cell Proliferation by the Cannabinoid Receptors CB1 and CB2 in Vascular Smooth Muscle Cells and Cardiac Myocytes. Biomedicines 2022; 10:biomedicines10123271. [PMID: 36552027 PMCID: PMC9775096 DOI: 10.3390/biomedicines10123271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cannabinoids (CB) are implicated in cardiovascular diseases via the two main receptor subtypes CB1R and CB2R. This study investigated whether cannabinoids regulate the activity of matrix metalloproteases (MMP-2, MMP-9) in vascular smooth muscle cells (VSMCs) and in cells of cardiac origin (H9c2 cell line). The influence of CB1- and CB2 receptor stimulation or inhibition on cell proliferation, apoptosis and glucose uptake was also evaluated. We used four compounds that activate or block CB receptors: arachidonyl-2-chloroethylamide (ACEA)-CB1R agonist, rimonabant-CB1R antagonist, John W. Huffman (JWH133)-CB2R agonist and CB2R antagonist-6-Iodopravadoline (AM630). Treatment of cells with the CB2R agonist JWH133 decreased cytokine activated secretion of proMMP-2, MMP-2 and MMP-9, reduced Fas ligand and caspase-3-mediated apoptosis, normalized the expression of TGF-beta1 and prevented cytokine-induced increase in glucose uptake into the cell. CB1R inhibition with rimonabant showed similar protective properties as the CB2R agonist JWH133, but to a lesser extent. In conclusion, CB1R and CB2R exert opposite effects on cell glucose uptake, proteolysis and apoptosis in both VSMCs and H9c2 cells. The CB2R agonist JWH133 demonstrated the highest protective properties. These findings may pave the way to a new treatment of cardiovascular diseases, especially those associated with extracellular matrix degradation.
Collapse
|
138
|
Zhou H, Liu S, Zhang N, Fang K, Zong J, An Y, Chang X. Downregulation of Sirt6 by CD38 promotes cell senescence and aging. Aging (Albany NY) 2022; 14:9730-9757. [PMID: 36490326 PMCID: PMC9792202 DOI: 10.18632/aging.204425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
Decreased nicotinamide adenine dinucleotide (NAD+) levels accompany aging. CD38 is the main cellular NADase. Cyanidin-3-O-glucoside (C3G), a natural inhibitor of CD38, is a well-known drug that extends the human lifespan. We investigated mechanisms of CD38 in cell senescence and C3G in antiaging. Myocardial H9c2 cells were induced to senescence with D-gal. CD38 siRNA, C3G and UBCS039 (a chemical activator of Sirt6) inhibited D-gal-induced senescence by reducing reactive oxygen species, hexokinase 2 and SA-β-galactosidase levels. These activators also stimulated cell proliferation and telomerase reverse transcriptase levels, while OSS-128167 (a chemical inhibitor of Sirt6) and Sirt6 siRNA exacerbated the senescent process. H9c2 cells that underwent D-gal-induced cell senescence increased CD38 expression and decreased Sirt6 expression; CD38 siRNA and C3G decreased CD38 expression and increased Sirt6 expression, respectively; and Sirt6 siRNA stimulated cell senescence in the presence of C3G and CD38 siRNA. In D-gal-induced acute aging mice, CD38 and Sirt6 exhibited increased and decreased expression, respectively, in myocardial tissues, and C3G treatment decreased CD38 expression and increased Sirt6 expression in the tissues. C3G also reduced IL-1β, IL-6, IL-17A, TNF-α levels and restored NAD+ and NK cell levels in the animals. We suggest that CD38 downregulates Sirt6 expression to promote cell senescence and C3G exerts an antiaging effect through CD38-Sirt6 signaling.
Collapse
Affiliation(s)
- Hongji Zhou
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China,Department of Cardiovascular Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Shihai Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - NanYang Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Kehua Fang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jinbao Zong
- Clinical Laboratory and Central Laboratory, The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Yi An
- Department of Cardiovascular Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xiaotian Chang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
139
|
Bax M, Romanov V, Junday K, Giannoulatou E, Martinac B, Kovacic JC, Liu R, Iismaa SE, Graham RM. Arterial dissections: Common features and new perspectives. Front Cardiovasc Med 2022; 9:1055862. [PMID: 36561772 PMCID: PMC9763901 DOI: 10.3389/fcvm.2022.1055862] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Arterial dissections, which involve an abrupt tear in the wall of a major artery resulting in the intramural accumulation of blood, are a family of catastrophic disorders causing major, potentially fatal sequelae. Involving diverse vascular beds, including the aorta or coronary, cervical, pulmonary, and visceral arteries, each type of dissection is devastating in its own way. Traditionally they have been studied in isolation, rather than collectively, owing largely to the distinct clinical consequences of dissections in different anatomical locations - such as stroke, myocardial infarction, and renal failure. Here, we review the shared and unique features of these arteriopathies to provide a better understanding of this family of disorders. Arterial dissections occur commonly in the young to middle-aged, and often in conjunction with hypertension and/or migraine; the latter suggesting they are part of a generalized vasculopathy. Genetic studies as well as cellular and molecular investigations of arterial dissections reveal striking similarities between dissection types, particularly their pathophysiology, which includes the presence or absence of an intimal tear and vasa vasorum dysfunction as a cause of intramural hemorrhage. Pathway perturbations common to all types of dissections include disruption of TGF-β signaling, the extracellular matrix, the cytoskeleton or metabolism, as evidenced by the finding of mutations in critical genes regulating these processes, including LRP1, collagen genes, fibrillin and TGF-β receptors, or their coupled pathways. Perturbances in these connected signaling pathways contribute to phenotype switching in endothelial and vascular smooth muscle cells of the affected artery, in which their physiological quiescent state is lost and replaced by a proliferative activated phenotype. Of interest, dissections in various anatomical locations are associated with distinct sex and age predilections, suggesting involvement of gene and environment interactions in disease pathogenesis. Importantly, these cellular mechanisms are potentially therapeutically targetable. Consideration of arterial dissections as a collective pathology allows insight from the better characterized dissection types, such as that involving the thoracic aorta, to be leveraged to inform the less common forms of dissections, including the potential to apply known therapeutic interventions already clinically available for the former.
Collapse
Affiliation(s)
- Monique Bax
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Valentin Romanov
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Keerat Junday
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Jason C. Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, United States
| | - Renjing Liu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Siiri E. Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
| | - Robert M. Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- UNSW Medicine and Health, UNSW Sydney, Kensington, NSW, Australia
- St. Vincent’s Hospital, Darlinghurst, NSW, Australia
| |
Collapse
|
140
|
Li X, Chen H, Zhang D. Discoidin domain receptor 1 may be involved in biological barrier homeostasis. J Clin Pharm Ther 2022; 47:2397-2407. [PMID: 35665520 DOI: 10.1111/jcpt.13705] [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: 12/04/2021] [Revised: 04/08/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase involved in the pathological processes of several diseases, such as keloid formation, renal fibrosis, atherosclerosis, tumours, and inflammatory processes. The biological barrier is the first line of defence against pathogens, and its disruption is closely related to diseases. In this review, we attempt to elucidate the relationship between DDR1 and the biological barrier, explore the potential biological value of DDR1, and review the current research status and clinical potential of DDR1-selective inhibitors. METHODS We conducted an extensive literature search on PubMed to collect studies on the relevance of DDR1 to biological barriers and DDR1-selective inhibitors. With these studies, we explored the relationship between DDR1 and biological barriers and briefly reviewed representative DDR1-selective inhibitors that have been reported in recent years. RESULTS AND DISCUSSION First, the review of the potential mechanisms by which DDR1 regulates biological barriers, including the epithelial, vascular, glomerular filtration, blood-labyrinth, and blood-brain barriers. In the body, DDR1 dysfunction and aberrant expression may be involved in the homeostasis of the biological barrier. Secondly, the review of DDR1 inhibitors reported in recent years shows that DDR1-targeted inhibition is an attractive and promising pharmacological intervention. WHAT IS NEW AND CONCLUSIONS This review shows that DDR1 is involved in various physiological and pathological processes and in the regulation of biological barrier homeostasis. However, studies on DDR1 and biological barriers are still scarce, and further studies are needed to elucidate their specific mechanisms. The development of targeted inhibitors provides a new direction and idea to study the mechanism of DDR1.
Collapse
Affiliation(s)
- Xiaoli Li
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Huiling Chen
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Department of Gastroenterology, Key Laboratory of Digestive Diseases, LanZhou University Second Hospital, LanZhou University, Lanzhou, China
| |
Collapse
|
141
|
Wu M, Liu W, Huang H, Chen Z, Chen Y, Zhong Y, Jin Z, Liu X, Zou L. PVT1/miR-145-5p/HK2 modulates vascular smooth muscle cells phenotype switch via glycolysis: The new perspective on the spiral artery remodeling. Placenta 2022; 130:25-33. [PMID: 36370492 DOI: 10.1016/j.placenta.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/01/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Vascular smooth muscle cells (VSMC) switched from a contractile phenotype to a synthetic phenotype during the decidual spiral artery (SPAs) remodeling process. The lncRNA plasmacytoma variant translocation 1 (PVT1) and glucose metabolism have been found to regulate the VSMC phenotype switch. This study aimed to analyze the dynamic expression of PVT1 and glycolytic key enzymes hexokinase2 (HK2) at different remodeling stages in early human pregnancy and elucidate the underlying mechanism of the PVT1/miR-145-5p/HK2 axis involved in the spiral artery remodeling. METHODS qRT-PCR, Western blot (WB) analysis, Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) were used to detect the expression and localization of PVT1 and HK2 in decidual tissue. HA-VSMCs were transfected with specific siRNA, shRNA and plasmids to regulate corresponding genes. Extracellular lactate, cellular ATP, ROS, and intracellular NADPH levels were measured using the corresponding assay kits. Migration was measured by wound-healing and Transwell assays. Contractile phenotypic markers α-SMA, MYH11 with calponin and synthetic phenotypic markers OPN and vimentin were detected by WB. The PDC model was used to detect the degree of spiral arterial remodeling. RESULTS PVT1 and HK2 were upregulated with gestational age (GA) increasing in decidual tissue during the early pregnancy. HK2 regulated the glycolytic activity and VSMC phenotype switch in vitro. PVT1 regulated the glycolytic activity and VSMC phenotype switch through HK2. PVT1 played a ceRNA role in regulating HK2 expression by sponging miR-145-5p. PVT1 and HK2 influenced spiral artery remodeling in the PDC model. DISCUSSION PVT1 and HK2 were upregulated, and miR-145-5p was downregulated in decidua with the GA increasing. Meanwhile, the PVT1/miR-145-5p/HK2 axis may be involved in regulating the phenotypic switch and migratory capacity of VSMCs by affecting glycolysis in decidual SPAs remodeling.
Collapse
Affiliation(s)
- Mengying Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weifang Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haixia Huang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhirui Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangyang Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqi Zhong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhishan Jin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
142
|
Patil N, Howe O, Cahill P, Byrne HJ. Monitoring and modelling the dynamics of the cellular glycolysis pathway: A review and future perspectives. Mol Metab 2022; 66:101635. [PMID: 36379354 PMCID: PMC9703637 DOI: 10.1016/j.molmet.2022.101635] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The dynamics of the cellular glycolysis pathway underpin cellular function and dysfunction, and therefore ultimately health, disease, diagnostic and therapeutic strategies. Evolving our understanding of this fundamental process and its dynamics remains critical. SCOPE OF REVIEW This paper reviews the medical relevance of glycolytic pathway in depth and explores the current state of the art for monitoring and modelling the dynamics of the process. The future perspectives of label free, vibrational microspectroscopic techniques to overcome the limitations of the current approaches are considered. MAJOR CONCLUSIONS Vibrational microspectroscopic techniques can potentially operate in the niche area of limitations of other omics technologies for non-destructive, real-time, in vivo label-free monitoring of glycolysis dynamics at a cellular and subcellular level.
Collapse
Affiliation(s)
- Nitin Patil
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland; School of Physics and Optometric & Clinical Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland.
| | - Orla Howe
- School of Biological and Health Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland
| | - Paul Cahill
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Hugh J Byrne
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland
| |
Collapse
|
143
|
Mokry M, Boltjes A, Slenders L, Bel-Bordes G, Cui K, Brouwer E, Mekke JM, Depuydt MA, Timmerman N, Waissi F, Verwer MC, Turner AW, Khan MD, Hodonsky CJ, Benavente ED, Hartman RJ, van den Dungen NAM, Lansu N, Nagyova E, Prange KH, Kovacic JC, Björkegren JL, Pavlos E, Andreakos E, Schunkert H, Owens GK, Monaco C, Finn AV, Virmani R, Leeper NJ, de Winther MP, Kuiper J, de Borst GJ, Stroes ES, Civelek M, de Kleijn DP, den Ruijter HM, Asselbergs FW, van der Laan SW, Miller CL, Pasterkamp G. Transcriptomic-based clustering of human atherosclerotic plaques identifies subgroups with different underlying biology and clinical presentation. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1140-1155. [PMID: 37920851 PMCID: PMC10621615 DOI: 10.1038/s44161-022-00171-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/20/2022] [Indexed: 11/04/2023]
Abstract
Histopathological studies have revealed key processes of atherosclerotic plaque thrombosis. However, the diversity and complexity of lesion types highlight the need for improved sub-phenotyping. Here we analyze the gene expression profiles of 654 advanced human carotid plaques. The unsupervised, transcriptome-driven clustering revealed five dominant plaque types. These plaque phenotypes were associated with clinical presentation and showed differences in cellular compositions. Validation in coronary segments showed that the molecular signature of these plaques was linked to coronary ischemia. One of the plaque types with the most severe clinical symptoms pointed to both inflammatory and fibrotic cell lineages. Further, we did a preliminary analysis of potential circulating biomarkers that mark the different plaques phenotypes. In conclusion, the definition of the plaque at risk for a thrombotic event can be fine-tuned by in-depth transcriptomic-based phenotyping. These differential plaque phenotypes prove clinically relevant for both carotid and coronary artery plaques and point to distinct underlying biology of symptomatic lesions.
Collapse
Affiliation(s)
- Michal Mokry
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Arjan Boltjes
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Gemma Bel-Bordes
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Kai Cui
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Eli Brouwer
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Joost M. Mekke
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Marie A.C. Depuydt
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Nathalie Timmerman
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Farahnaz Waissi
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Maarten C Verwer
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Mohammad Daud Khan
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Chani J. Hodonsky
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Ernest Diez Benavente
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Robin J.G. Hartman
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Noortje A M van den Dungen
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Nico Lansu
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Emilia Nagyova
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Koen H.M. Prange
- Amsterdam University Medical Centers – location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - Jason C. Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and St Vincent’s Clinical School, University of New South Wales, Australia
| | - Johan L.M. Björkegren
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Eleftherios Pavlos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), Partner Site Munich Heart Alliance, Munich, Germany
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford
| | | | | | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA
| | - Menno P.J. de Winther
- Amsterdam University Medical Centers – location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Gert J. de Borst
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | | | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, United Kingdom
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
144
|
Li D, Guo J, Ni X, Sun G, Bao H. The progress and challenges of circRNA for diabetic foot ulcers: A mini-review. Front Endocrinol (Lausanne) 2022; 13:1019935. [PMID: 36531481 PMCID: PMC9747764 DOI: 10.3389/fendo.2022.1019935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
Since the Human Genome Project was successfully completed, humanity has entered a post-genome era, and the second-generation sequencing technology has gradually progressed and become more accurate. Meanwhile, circRNAs plays a crucial role in the regulation of diseases and potential clinical applications has gradually attracted the attention of physicians. However, the mechanisms of circRNAs regulation at the cellular and molecular level of diabetic foot ulcer (DFU) is still not well-understood. With the deepening of research, there have been many recent studies conducted to explore the effect of circRNAs on DFU. In this mini-review, we discuss the potential role of circRNAs as therapeutic targets and diagnostic markers for DFU in order to gain a better understanding of the molecular mechanisms that underlie the development of DFU and to establish a theoretical basis for accurate treatment and effective prevention.
Collapse
Affiliation(s)
- Deer Li
- Graduate School, Inner Mongolia Medical University, Hohhot, China
- Department of Traumatology and Orthopedics, Inner Mongolia People’s Hospital, Hohhot, China
| | - Jiaxing Guo
- Department of Joint Surgery, The Second Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| | - Xiyu Ni
- Graduate School, Inner Mongolia Medical University, Hohhot, China
- Department of Traumatology and Orthopedics, Inner Mongolia People’s Hospital, Hohhot, China
| | - Guanwen Sun
- Department of Traumatology and Orthopedics, Inner Mongolia People’s Hospital, Hohhot, China
| | - Huhe Bao
- Department of Traumatology and Orthopedics, Inner Mongolia People’s Hospital, Hohhot, China
| |
Collapse
|
145
|
Sun LY, Lyu YY, Zhang HY, Shen Z, Lin GQ, Geng N, Wang YL, Huang L, Feng ZH, Guo X, Lin N, Ding S, Yuan AC, Zhang L, Qian K, Pu J. Nuclear Receptor NR1D1 Regulates Abdominal Aortic Aneurysm Development by Targeting the Mitochondrial Tricarboxylic Acid Cycle Enzyme Aconitase-2. Circulation 2022; 146:1591-1609. [PMID: 35880522 PMCID: PMC9674448 DOI: 10.1161/circulationaha.121.057623] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 06/10/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Metabolic disorder increases the risk of abdominal aortic aneurysm (AAA). NRs (nuclear receptors) have been increasingly recognized as important regulators of cell metabolism. However, the role of NRs in AAA development remains largely unknown. METHODS We analyzed the expression profile of the NR superfamily in AAA tissues and identified NR1D1 (NR subfamily 1 group D member 1) as the most highly upregulated NR in AAA tissues. To examine the role of NR1D1 in AAA formation, we used vascular smooth muscle cell (VSMC)-specific, endothelial cell-specific, and myeloid cell-specific conditional Nr1d1 knockout mice in both AngII (angiotensin II)- and CaPO4-induced AAA models. RESULTS Nr1d1 gene expression exhibited the highest fold change among all 49 NRs in AAA tissues, and NR1D1 protein was upregulated in both human and murine VSMCs from AAA tissues. The knockout of Nr1d1 in VSMCs but not endothelial cells and myeloid cells inhibited AAA formation in both AngII- and CaPO4-induced AAA models. Mechanistic studies identified ACO2 (aconitase-2), a key enzyme of the mitochondrial tricarboxylic acid cycle, as a direct target trans-repressed by NR1D1 that mediated the regulatory effects of NR1D1 on mitochondrial metabolism. NR1D1 deficiency restored the ACO2 dysregulation and mitochondrial dysfunction at the early stage of AngII infusion before AAA formation. Supplementation with αKG (α-ketoglutarate, a downstream metabolite of ACO2) was beneficial in preventing and treating AAA in mice in a manner that required NR1D1 in VSMCs. CONCLUSIONS Our data define a previously unrecognized role of nuclear receptor NR1D1 in AAA pathogenesis and an undescribed NR1D1-ACO2 axis involved in regulating mitochondrial metabolism in VSMCs. It is important that our findings suggest αKG supplementation as an effective therapeutic approach for AAA treatment.
Collapse
MESH Headings
- Humans
- Mice
- Animals
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/prevention & control
- Aorta, Abdominal/pathology
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Muscle, Smooth, Vascular/metabolism
- Citric Acid Cycle
- Myocytes, Smooth Muscle/metabolism
- Angiotensin II/adverse effects
- Mice, Knockout
- Aconitate Hydratase/metabolism
- Disease Models, Animal
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Ling-Yue Sun
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Yan Lyu
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Heng-Yuan Zhang
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Shen
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Guan-Qiao Lin
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Na Geng
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Li Wang
- Department of Vascular Surgery (Y.-L.W., L.Z.), Shanghai Jiao Tong University, Shanghai, China
| | - Lin Huang
- Renji Hospital, School of Medicine, School of Biomedical Engineering and Med-X Research Institute (L.H., K.Q.), Shanghai Jiao Tong University, Shanghai, China
| | - Ze-Hao Feng
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Guo
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Nan Lin
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Song Ding
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - An-Cai Yuan
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Lan Zhang
- Department of Vascular Surgery (Y.-L.W., L.Z.), Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Renji Hospital, School of Medicine, School of Biomedical Engineering and Med-X Research Institute (L.H., K.Q.), Shanghai Jiao Tong University, Shanghai, China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
146
|
Fan D, Liu C, Zhang Z, Huang K, Wang T, Chen S, Li Z. Progress in the Preclinical and Clinical Study of Resveratrol for Vascular Metabolic Disease. Molecules 2022; 27:7524. [PMID: 36364370 PMCID: PMC9658204 DOI: 10.3390/molecules27217524] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Vascular metabolic dysfunction presents in various diseases, such as atherosclerosis, hypertension, and diabetes mellitus. Due to the high prevalence of these diseases, it is important to explore treatment strategies to protect vascular function. Resveratrol (RSV), a natural polyphenolic phytochemical, is regarded as an agent to regulate metabolic pathways. Many studies have proven that RSV has beneficial effects on improving metabolism in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), which provide new directions to treat vascular metabolic diseases. Herein, we overviewed that RSV could regulate cell metabolism activity by inhibiting glucose uptake, suppressing glycolysis, preventing cells from fatty acid-related damages, reducing lipogenesis, increasing fatty acid oxidation, enhancing lipolysis, elevating uptake and synthesis of glutamine, and increasing NO release. Furthermore, in clinical trials, although the results from different studies remain controversial, we proposed that RSV had better therapeutic effects at high concentrations and for patients with metabolic disorders.
Collapse
Affiliation(s)
- Dongxiao Fan
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Chenshu Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhongyu Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kan Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
147
|
Li H, Xu J, Zhang Y, Hong L, He Z, Zeng Z, Zhang L. Astragaloside IV alleviates senescence of vascular smooth muscle cells through activating Parkin-mediated mitophagy. Hum Cell 2022; 35:1684-1696. [PMID: 35925474 PMCID: PMC9515037 DOI: 10.1007/s13577-022-00758-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023]
Abstract
Astragaloside IV (AS-IV), as one of the main active components of Astragalus membranaceus, has been reported to have cardiovascular protective effects. However, the role and molecular mechanism of AS-IV in vascular senescence have not been clearly stated. The in vitro aging model was constructed using bleomycin (BLM) in vascular smooth muscle cells (VSMCs). Cell senescence were assessed through Western blotting analysis of aging markers, flow cytometry, and the β-galactosidase (SA-β-Gal) kit. Mitophagy was determined through transmission electron microscopy, TMRM staining, and Western blotting analysis of p62. A model of aging blood vessels was induced by D-gal. The vascular wall thickness of mice was also evaluated by H&E staining. Our data proved that AS-IV plays an anti-senescent role in vitro and in vivo. Results showed that AS-IV effectively improved mitochondrial injury, raised MMP, and mediated mitophagy in BLM-induced senescent VSMCs and D-gal induced aging mice. Parkin expression strengthened AS-IV's anti-senescent function. In conclusions, AS-IV attenuated BLM-induced VSMC senescence via Parkin to regulate mitophagy. Therefore, AS-IV-mediated Parkin might be a latent therapeutic agent and target for VSMC senescence.
Collapse
Affiliation(s)
- Huijun Li
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19, Nonglinxia Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Jialin Xu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19, Nonglinxia Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Yanan Zhang
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Lei Hong
- Department of Cardiology, Long Gang Central Hospital of Shenzhen, Shenzhen, 518116, Guangdong, China
| | - Zhijian He
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19, Nonglinxia Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Zhiheng Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19, Nonglinxia Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19, Nonglinxia Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
148
|
Affiliation(s)
- Kathy O Lui
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China (K.O.L.)
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China (Y.H.)
| |
Collapse
|
149
|
Jia Y, Mao C, Ma Z, Huang J, Li W, Ma X, Zhang S, Li M, Yu F, Sun Y, Chen J, Feng J, Zhou Y, Xu Q, Zhao L, Fu Y, Kong W. PHB2 Maintains the Contractile Phenotype of VSMCs by Counteracting PKM2 Splicing. Circ Res 2022; 131:807-824. [PMID: 36200440 DOI: 10.1161/circresaha.122.321005] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Phenotypic transition of vascular smooth muscle cells (VSMCs) accounts for the pathogenesis of a variety of vascular diseases during the early stage. Recent studies indicate the metabolic reprogramming may be involved in VSMC phenotypic transition. However, the definite molecules that link energy metabolism to distinct VSMC phenotype remain elusive. METHODS A carotid artery injury model was used to study postinjury neointima formation as well as VSMC phenotypic transition in vivo. RNA-seq analysis, cell migration assay, collagen gel contraction assay, wire myography assay, immunoblotting, protein interactome analysis, co-immunoprecipitation, and mammalian 2-hybrid assay were performed to clarify the phenotype and elucidate the molecular mechanisms. RESULTS We collected cell energy-regulating genes by using Gene Ontology annotation and applied RNA-Seq analysis of transforming growth factor-β or platelet-derived growth factor BB stimulated VSMCs. Six candidate genes were overlapped from energy metabolism-related genes and genes reciprocally upregulated by transforming growth factor-β and downregulated by platelet-derived growth factor BB. Among them, prohibitin 2 has been reported to regulate mitochondrial oxidative phosphorylation. Indeed, prohibitin 2-deficient VSMCs lost the contractile phenotype as evidenced by reduced contractile proteins. Consistently, Phb2SMCKO mice were more susceptible to postinjury VSMC proliferation and neointima formation compared with Phb2flox/flox mice. Further protein interactome analysis, co-immunoprecipitation, and mammalian 2-hybrid assay revealed that prohibitin 2, through its C-terminus, directly interacts with hnRNPA1, a key modulator of pyruvate kinase M1/2 (PKM) mRNA splicing that promotes PKM2 expression and glycolysis. Prohibitin 2 deficiency facilitated PKM1/2 mRNA splicing and reversion from PKM1 to PKM2, and enhanced glycolysis in VSMCs. Blocking prohibitin 2-hnRNPA1 interaction resulted in increased PKM2 expression, enhanced glycolysis, repressed contractile marker genes expression in VSMCs, as well as aggravated postinjury neointima formation in vivo. CONCLUSIONS Prohibitin 2 maintains VSMC contractile phenotype by interacting with hnRNPA1 to counteract hnRNPA1-mediated PKM alternative splicing and glucose metabolic reprogramming.
Collapse
Affiliation(s)
- Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Chenfeng Mao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.).,Beijing Institute of Biotechnology, Beijing, P. R. China (C.M.)
| | - Zihan Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Xiaolong Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Siting Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Meihong Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Yingying Sun
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China (Y.S., J.C.)
| | - Jingzhou Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China (Y.S., J.C.)
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Yuan Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Qingbo Xu
- Cardiovascular Division, Kings College London BHF Centre, London SE5 9NU, UK (Q.X.).,Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China (Q.X.)
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, P. R. China (L.Z.)
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.J., C.M., Z.M., J.H., W.L., X.M., S.Z., M.L., F.Y., J.F., Y.Z., Y.F., W.K.)
| |
Collapse
|
150
|
Zhang J, Zhao WR, Shi WT, Tan JJ, Zhang KY, Tang JY, Chen XL, Zhou ZY. Tribulus terrestris L. extract ameliorates atherosclerosis by inhibition of vascular smooth muscle cell proliferation in ApoE -/- mice and A7r5 cells via suppression of Akt/MEK/ERK signaling. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115547. [PMID: 35870688 DOI: 10.1016/j.jep.2022.115547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/03/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis (AS) is one of major threatens of death worldwide, and vascular smooth muscle cell (VSMC) proliferation is an important characteristic in the progression of AS. Tribulus terrestris L. is a well-known Chinese Materia Medica for treating skin pruritus, vertigo and cardiovascular diseases in traditional Chinese medicine. However, its anti-AS activity and inhibition effect on VSMC proliferation are not fully elucidated. AIMS We hypothesize that the furostanol saponins enriched extract (FSEE) of T. terrestris L. presents anti-AS effect by inhibition of VSMC proliferation. The molecular action mechanism underlying the anti-VSMC proliferation effect of FSEE is also investigated. MATERIALS AND METHODS Apolipoprotein-E deficient (ApoE-/-) mice and rat thoracic smooth muscle cell A7r5 were employed as the in vivo and in vitro models respectively to evaluate the anti- AS and VSMC proliferation effects of FSEE. In ApoE-/- mice, the amounts of total cholesterol, triglyceride, low density lipoprotein and high density lipoprotein in serum were measured by commercially available kits. The size of atherosclerotic plaque was observed by hematoxylin & eosin staining. The protein expressions of α-smooth muscle actin (α-SMA) and osteopontin (OPN) in the plaque were examined by immunohistochemistry. In A7r5 cells, the cell viability and proliferation were tested by MTT and Real Time Cell Analysis assays. The cell migration was evaluated by wound healing assay. Propidium iodide staining followed by flow cytometry was used to analyze the cell cycle progression. The expression of intracellular total and phosphorylated proteins including protein kinase B (Akt) and mitogen-activated protein kinases (MAPKs), such as mitogen-activated extracellular signal-regulated kinase (MEK), extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), were detected by western blotting analysis. RESULTS FSEE significantly reduced the area of atherosclerotic plaque in high-fat diet-fed ApoE-/- mice. And FSEE increased the protein expression level of α-SMA and decreased the level of OPN in atherosclerotic plaque, which revealed the inhibition of VSMC phenotype switching and proliferation. In A7r5 cells, FSEE suppressed fetal bovine serum (FBS) or oxidized low density lipoprotein (oxLDL)-triggered VSMC proliferation and migration in a concentration dependent manner. FSEE protected against the elevation of cell numbers in S phase induced by FBS or oxLDL and the reduction of cell numbers in G0/G1 phase induced by oxLDL. Moreover, the phosphorylation of Akt and MAPKs including MEK, ERK and JNK could be facilitated by FBS or oxLDL, while co-treatment of FSEE attenuated the phosphorylation of Akt induced by oxLDL as well as the phosphorylation of MEK and ERK induced by FBS. In addition, (25R)-terrestrinin B (JL-6), which was the main ingredient of FSEE, and its potential active pharmaceutical ingredients tigogenin (Tigo) and hecogenin (Heco) also significantly attenuated FBS or oxLDL-induced VSMC proliferation in A7r5 cells. CONCLUSION FSEE presents potent anti- AS and VSMC proliferation activities and the underlying mechanism is likely to the suppression of Akt/MEK/ERK signaling. The active components of FSEE are JL-6 and its potential active pharmaceutical ingredients Tigo and Heco. So, FSEE and its active compounds may be potential therapeutic drug candidates for AS.
Collapse
Affiliation(s)
- Jing Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wai-Rong Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wen-Ting Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jun-Jie Tan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Kai-Yu Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jing-Yi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xin-Lin Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhong-Yan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|