201
|
Wiercigroch E, Stepula E, Mateuszuk L, Zhang Y, Baranska M, Chlopicki S, Schlücker S, Malek K. ImmunoSERS microscopy for the detection of smooth muscle cells in atherosclerotic plaques. Biosens Bioelectron 2019; 133:79-85. [PMID: 30909016 DOI: 10.1016/j.bios.2019.02.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/19/2019] [Accepted: 02/26/2019] [Indexed: 01/19/2023]
Abstract
We investigated the suitability of immuno-SERS (iSERS) microscopy for imaging of smooth muscle cells (SMCs) in atherosclerotic plaques. Localization of SMCs is achieved by using SERS-labelled antibodies direct against alpha-smooth muscle actin (SMA). The staining quality of the false-colour iSERS images obtained by confocal Raman microscopy with point mapping is compared with wide-field immunofluorescence images. Both direct (labelled primary antibody) and indirect iSERS staining (unlabelled primary and labelled secondary antibody) techniques were employed. Direct iSERS staining yields results comparable to indirect IF staining, demonstrating the suitability of iSERS in research on atherosclerosis and paving the way for future multiplexed imaging experiments.
Collapse
Affiliation(s)
- Ewelina Wiercigroch
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, Krakow 30-387, Poland; Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Elzbieta Stepula
- Department of Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstr. 5, 45141 Essen, Germany
| | - Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Yuying Zhang
- Department of Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstr. 5, 45141 Essen, Germany
| | - Malgorzata Baranska
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, Krakow 30-387, Poland; Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Chair of Pharmacology, Jagiellonian University, Medical College, Grzegorzecka 16, 31-531 Krakow, Poland
| | - Sebastian Schlücker
- Department of Chemistry and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstr. 5, 45141 Essen, Germany.
| | - Kamilla Malek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, Krakow 30-387, Poland; Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland.
| |
Collapse
|
202
|
Li HY, Leu YL, Wu YC, Wang SH. Melatonin Inhibits in Vitro Smooth Muscle Cell Inflammation and Proliferation and Atherosclerosis in Apolipoprotein E-Deficient Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:1889-1901. [PMID: 30661353 DOI: 10.1021/acs.jafc.8b06217] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Chronic inflammation and proliferation play important roles in atherosclerosis progression. This study aimed to identify the mechanisms responsible for the anti-inflammatory and antiproliferative effects of melatonin on tumor necrosis factor-α (TNF-α)- and platelet-derived growth factor-BB (PDGF-BB)-treated rat aortic smooth muscle cells (RASMCs). Melatonin reduced TNF-α-induced RASMC inflammation by decreasing vascular cell adhesion molecule-1 (VCAM-1) expression and nuclear factor-kappa B (NF-κB) P65 activity by inhibiting P38 mitogen-activated protein kinase phosphorylation ( P < 0.05). Additionally, melatonin inhibited PDGF-BB-induced RASMC proliferation by reducing mammalian target of rapamycin (mTOR) phosphorylation ( P < 0.05) but not migration in vitro. Melatonin also reduced TNF-α- and PDGF-BB-induced reactive oxygen species (ROS) production ( P < 0.05). Furthermore, melatonin treatment (prevention and treatment groups) significantly repressed high cholesterol diet-stimulated atherosclerotic lesions in vivo (19.59 ± 4.11%, 20.28 ± 5.63%, 32.26 ± 12.06%, respectively, P < 0.05). Taken together, the present study demonstrated that melatonin attenuated TNF-α-induced RASMC inflammation and PDGF-BB-induced RASMC proliferation in cells and reduced atherosclerotic lesions in mice. These results showed that melatonin has anti-inflammatory and antiproliferative properties and may be a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Hung-Yuan Li
- Department of Internal Medicine , National Taiwan University Hospital , Taipei , Taiwan
| | - Yann-Lii Leu
- Center for Traditional Chinese Medicine , Chang Gung Memorial Hospital , Taoyuan , Taiwan
| | - Ya-Chieh Wu
- Department of Nursing , Ching-Kuo Institute of Management and Health , Keelung , Taiwan
| | - Shu-Huei Wang
- Department of Anatomy and Cell Biology , College of Medicine, National Taiwan University , Taipei , Taiwan
| |
Collapse
|
203
|
Han Z, Yu Y, Xu J, Bao Z, Xu Z, Hu J, Yu M, Bamba D, Ma W, Ding F, Zhang L, Jin M, Yan G, Huang Q, Wang X, Hua B, Yang F, Li Y, Lei L, Cao N, Pan Z, Cai B. Iron Homeostasis Determines Fate of Human Pluripotent Stem Cells Via Glycerophospholipids-Epigenetic Circuit. Stem Cells 2019; 37:489-503. [PMID: 30599084 DOI: 10.1002/stem.2967] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/10/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022]
Abstract
Iron homeostasis is crucial for a variety of biological processes, but the biological role of iron homeostasis in pluripotent stem cells (PSCs) remains largely unknown. The present study aimed to determine whether iron homeostasis is involved in maintaining the pluripotency of human PSCs (hPSCs). We found that the intracellular depletion of iron leads to a rapid downregulation of NANOG and a dramatic decrease in the self-renewal of hPSCs as well as spontaneous and nonspecific differentiation. Moreover, long-term depletion of iron can result in the remarkable cell death of hPSCs via apoptosis and necrosis pathways. Additionally, we found that the depletion of iron increased the activity of lipoprotein-associated phospholipase A2 (LP-PLA2) and the production of lysophosphatidylcholine, thereby suppressing NANOG expression by enhancer of zeste homolog 2-mediated trimethylation of histone H3 lysine 27. Consistently, LP-PLA2 inhibition abrogated iron depletion-induced loss of pluripotency and differentiation. Altogether, the findings of our study demonstrates that iron homeostasis, acting through glycerophospholipid metabolic pathway, is essential for the pluripotency and survival of hPSCs. Stem Cells 2019;37:489-503.
Collapse
Affiliation(s)
- Zhenbo Han
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Ying Yu
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Juan Xu
- Department of Bioinformatics, Harbin Medical University, Harbin, People's Republic of China
| | - Zhengyi Bao
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Zihang Xu
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Jiancheng Hu
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Meixi Yu
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Djibril Bamba
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Wenya Ma
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Fengzhi Ding
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Lai Zhang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Mengyu Jin
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Gege Yan
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Qi Huang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Xiuxiu Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Bingjie Hua
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Fan Yang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Yuan Li
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Lei Lei
- Department of Histology and Embryology, Harbin Medical University, Harbin, People's Republic of China
| | - Nan Cao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.,The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People's Republic of China
| | - Zhenwei Pan
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Benzhi Cai
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
204
|
Rheumatoid Arthritis: Atherosclerosis Imaging and Cardiovascular Risk Assessment Using Machine and Deep Learning-Based Tissue Characterization. Curr Atheroscler Rep 2019; 21:7. [PMID: 30684090 DOI: 10.1007/s11883-019-0766-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF THE REVIEW Rheumatoid arthritis (RA) is a chronic, autoimmune disease which may result in a higher risk of cardiovascular (CV) events and stroke. Tissue characterization and risk stratification of patients with rheumatoid arthritis are a challenging problem. Risk stratification of RA patients using traditional risk factor-based calculators either underestimates or overestimates the CV risk. Advancements in medical imaging have facilitated early and accurate CV risk stratification compared to conventional cardiovascular risk calculators. RECENT FINDING In recent years, a link between carotid atherosclerosis and rheumatoid arthritis has been widely discussed by multiple studies. Imaging the carotid artery using 2-D ultrasound is a noninvasive, economic, and efficient imaging approach that provides an atherosclerotic plaque tissue-specific image. Such images can help to morphologically characterize the plaque type and accurately measure vital phenotypes such as media wall thickness and wall variability. Intelligence-based paradigms such as machine learning- and deep learning-based techniques not only automate the risk characterization process but also provide an accurate CV risk stratification for better management of RA patients. This review provides a brief understanding of the pathogenesis of RA and its association with carotid atherosclerosis imaged using the B-mode ultrasound technique. Lacunas in traditional risk scores and the role of machine learning-based tissue characterization algorithms are discussed and could facilitate cardiovascular risk assessment in RA patients. The key takeaway points from this review are the following: (i) inflammation is a common link between RA and atherosclerotic plaque buildup, (ii) carotid ultrasound is a better choice to characterize the atherosclerotic plaque tissues in RA patients, and (iii) intelligence-based paradigms are useful for accurate tissue characterization and risk stratification of RA patients.
Collapse
|
205
|
Mittal R, Jhaveri VM, Kay SIS, Greer A, Sutherland KJ, McMurry HS, Lin N, Mittal J, Malhotra AK, Patel AP. Recent Advances in Understanding the Pathogenesis of Cardiovascular Diseases and Development of Treatment Modalities. Cardiovasc Hematol Disord Drug Targets 2019; 19:19-32. [PMID: 29737266 DOI: 10.2174/1871529x18666180508111353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/15/2017] [Accepted: 03/28/2018] [Indexed: 06/08/2023]
Abstract
Cardiovascular Diseases (CVDs) are a leading cause of morbidity and mortality worldwide. The underlying pathology for cardiovascular disease is largely atherosclerotic in nature and the steps include fatty streak formation, plaque progression and plaque rupture. While there is optimal drug therapy available for patients with CVD, there are also underlying drug delivery obstacles that must be addressed. Challenges in drug delivery warrant further studies for the development of novel and more efficacious medical therapies. An extensive understanding of the molecular mechanisms of disease in combination with current challenges in drug delivery serves as a platform for the development of novel drug therapeutic targets for CVD. The objective of this article is to review the pathogenesis of atherosclerosis, first-line medical treatment for CVD, and key obstacles in an efficient drug delivery.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Vasanti M Jhaveri
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Sae-In Samantha Kay
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida FL, United States
| | - Aubrey Greer
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Kyle J Sutherland
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Hannah S McMurry
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Nicole Lin
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Arul K Malhotra
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Amit P Patel
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida FL, United States
| |
Collapse
|
206
|
Ng FL, Ong YO, Chen HZ, Tran LQN, Cao Y, Tay BY, Tan LP. A facile method for fabricating a three-dimensional aligned fibrous scaffold for vascular application. RSC Adv 2019; 9:13054-13064. [PMID: 35520779 PMCID: PMC9063778 DOI: 10.1039/c9ra00661c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/05/2019] [Indexed: 01/03/2023] Open
Abstract
Collection of circumferentially aligned and 3D fibrous scaffold on a newly designed electrospinning auxiliary jig. The aligned fibres served as a signaling modality to induce cell alignment and the maintenance of a contractile phenotype for hSMCs.
Collapse
Affiliation(s)
- Feng Lin Ng
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
- Singapore Institute of Manufacturing Technology
| | - Yee Oon Ong
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| | - Hui Zhi Chen
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| | - Le Quan Ngoc Tran
- Singapore Institute of Manufacturing Technology
- Singapore 637662
- Singapore
| | - Ye Cao
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| | - Bee Yen Tay
- Singapore Institute of Manufacturing Technology
- Singapore 637662
- Singapore
| | - Lay Poh Tan
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| |
Collapse
|
207
|
Wu L, Li Y, Zhang D, Huang Z, Du B, Wang Z, Yang L, Zhang Y. LncRNA NEXN-AS1 attenuates proliferation and migration of vascular smooth muscle cells through sponging miR-33a/b. RSC Adv 2019; 9:27856-27864. [PMID: 35530470 PMCID: PMC9070771 DOI: 10.1039/c9ra06282c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 08/27/2019] [Indexed: 12/27/2022] Open
Abstract
Non-protein-coding RNAs (lncRNAs) are emerging as important regulators in disease pathogenesis, including atherosclerosis (AS). Here, we investigated the role and underlying mechanisms of nexilin F-actin binding protein antisense RNA 1 (NEXN-AS1) on the proliferation and migration of vascular smooth muscle cells (VSMCs). Our data revealed that ox-LDL treatment resulted in decreased NEXN-AS1 expression and increased miR-33a/b levels in human aorta VSMCs (HA-VSMCs) in dose- and time-dependent manners. Overexpression of NEXN-AS1 mitigated the proliferation and migration of HA-VSMCs under ox-LDL stimulation using CCK-8 and wound-healing assays. Moreover, dual-luciferase reporter and RNA immunoprecipitation assays verified that NEXN-AS1 acted as molecular sponges of miR-33a and miR-33b in HA-VSMCs. MiR-33a or miR-33b silencing attenuated the proliferation and migration of ox-LDL-treated HA-VSMCs. Furthermore, miR-33a or miR-33b mediated the inhibitory effects of NEXN-AS1 overexpression on the proliferation and migration of ox-LDL-treated HA-VSMCs. Our study suggested that high level of NEXN-AS1 mitigated VSMC proliferation and migration under ox-LDL stimulation at least partly through sponging miR-33a and miR-33b, illuminating NEXN-AS1 as a novel therapeutic approach for AS treatment. Non-protein-coding RNAs (lncRNAs) are emerging as important regulators in disease pathogenesis, including atherosclerosis (AS).![]()
Collapse
Affiliation(s)
- Leiming Wu
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| | - Yapeng Li
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| | - Dianhong Zhang
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| | - Zhen Huang
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| | - Binbin Du
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| | - Zheng Wang
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| | - Lulu Yang
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| | - Yanzhou Zhang
- Department of Cardiology
- The First Affiliated Hospital of Zhengzhou University
- Zhengzhou 450052
- China
| |
Collapse
|
208
|
Reactive Oxygen Species from NADPH Oxidase and Mitochondria Participate in the Proliferation of Aortic Smooth Muscle Cells from a Model of Metabolic Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5835072. [PMID: 30671170 PMCID: PMC6323422 DOI: 10.1155/2018/5835072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/27/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
In metabolic diseases, the increased reactive oxygen species (ROS) represents one of the pathogenic mechanisms for vascular disease probably by promoting vascular smooth muscle cell (SMC) proliferation that contributes to the development of arterial remodeling and stenosis, hypertension, and atherosclerosis. Therefore, this work was undertaken to evaluate the participation of ROS from NADPH oxidase and mitochondria in the proliferation of SMCs from the aorta in a model of metabolic syndrome induced by sucrose feeding in rats. After 24 weeks, sucrose-fed (SF) rats develop hypertension, intra-abdominal obesity, hyperinsulinemia, and hyperleptinemia. In addition SMCs from SF rats had a higher growth rate and produce more ROS than control cells. The treatment of SMCs with DPI and apocynin to inhibit NADPH oxidase and with tempol to scavenge superoxide anion significantly blocked the proliferation of both SF and control cells suggesting the participation of NADPH oxidase as a source of superoxide anion. MitoTEMPO, which targets mitochondria within the cell, also significantly inhibited the proliferation of SMCs having a greater effect on cells from SF than from the control aorta. The higher rate of cell growth from the SF aorta is supported by the increased content of cyclophilin A and CD147, proteins involved in the mechanism of cell proliferation. In addition, caldesmon, α-actin, and phosphorylated myosin light chain, contractile phenotype proteins, were found significantly lower in SF cells in no confluent state and increased in confluent state but without difference between both cell types. Our results suggest that ROS from NADPH oxidase and mitochondria significantly participate in the difference found in the rate of cell growth between SF and control cells.
Collapse
|
209
|
SIRT7 Regulates the Vascular Smooth Muscle Cells Proliferation and Migration via Wnt/ β-Catenin Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4769596. [PMID: 30627559 PMCID: PMC6304541 DOI: 10.1155/2018/4769596] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/09/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022]
Abstract
A huge amount of evidence indicates that sirtuin 7 (SIRT7), a key mediator of many cellular activities, plays a crucial role in the pathogenesis of various diseases. However, little is known about the role of SIRT7 in atherosclerosis. This study investigated the potential role of SIRT7 in regulating the proliferation and migration of human vascular smooth muscle cells (HAVSMCs) and its possible molecular mechanism. In this study, human vascular smooth muscle cells (HAVSMCs) were induced by oxidized low-density lipoprotein (ox-LDL) to establish atherosclerosis (AS) cell model. Immunofluorescence staining and Western blot were used to detect the level of α-SMA expression, which was a marker protein in AS. In addition, RT-qPCR and Western blot assay were applied for exploring the mRNA and protein expression levels of SIRT7, Wnt, β-catenin, and cyclin D1 after knockdown or overexpression of SIRT7. And, furthermore, Cell Counting Kit-8 assay, flow cytometry, and wound-healing assay were used to assess HAVSMCs proliferation, cell cycle, and migration. Dickkopf-1 (DKK-1), a secretory glycoprotein that can block Wnt/β-catenin pathway, was used in SIRT7 overexpression HAVSMCs; subsequently cells proliferation and migration were assessed by Cell Counting Kit-8 assay, flow cytometry analysis, and wound-healing assay. We found that knockdown of SIRT7 significantly promoted cell proliferation and migration, decreased the percentages of cells in the G1 and G2 phases, and increased those in the S phase and downregulated the protein expression levels of Wnt, β-catenin, and cyclin D1, while overexpression of SIRT7 had reverse results. After treatment with Wnt/beta-catenin pathway inhibitor DKK-1 in SIRT7 overexpression HAVSMCs, cell proliferation and migration were increased, respectively. In conclusion, SIRT7 inhibited HAVSMCs proliferation and migration via enhancing Wnt/β-catenin activation, which provided a novel therapeutic strategy for antiatherosclerosis.
Collapse
|
210
|
Liu Y, Jia L, Min D, Xu Y, Zhu J, Sun Z. Baicalin inhibits proliferation and promotes apoptosis of vascular smooth muscle cells by regulating the MEG3/p53 pathway following treatment with ox‑LDL. Int J Mol Med 2018; 43:901-913. [PMID: 30535498 PMCID: PMC6317676 DOI: 10.3892/ijmm.2018.4009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/26/2018] [Indexed: 11/15/2022] Open
Abstract
Atherosclerosis (AS) is a systemic disease associated with lipid metabolic disorders and abnormal proliferation of smooth muscle cells. Baicalin is a flavonoid compound isolated from the dry roots of Scutellaria baicalensis Georgi and exerts anti-proliferative effects in various types of cells. However, the effect of baicalin on AS remains unclear. In the present study, serum samples were collected from patients with AS and an in vitro model of AS was established using oxidized low-density lipoprotein (ox-LDL)-treated human aorta vascular smooth muscle cells (HA-VSMCs). The siRNA transfection and overexpression efficiency of endogenous maternally expressed gene 3 (MEG3) and the expression level of MEG3 were analyzed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The effects of alterations in expression levels of MEG3 were assessed by MTT assay, bromodeoxyuridine incorporation assay, 5-ethynyl-2′-deoxyuridine staining, wound healing assay, immunofluorescence and western blotting in HA-VSMCs. qPCR indicated that the expression of MEG3 was reduced in serum samples from patients with AS and ox-LDL-treated HA-VSMCs, compared with serum samples from healthy patients and untreated HA-VSMCs, respectively. Further experiments indicated that ox-LDL-induced decrease of MEG3 expression was reversed by treatment with baicalin in a concentration-dependent manner. Following treatment with ox-LDL, decreased expression of MEG3 promoted proliferation and migration, and suppressed apoptosis in HA-VSMCs. Furthermore, treatment with baicalin reversed these effects on proliferation and apoptosis in ox-LDL-treated HA-VSMCs. The current study indicated that downregulated expression of MEG3 increased cell cycle-associated protein expression. However, treatment with baicalin inhibited the expression of cell-cycle associated proteins in HA-VSMCs with MEG3 knockdown. In addition, baicalin activated the p53 signaling pathway and promoted the expression and transport of p53 from the cytoplasm to nucleus following MEG3 knockdown in ox-LDL-treated HA-VSMCs. Baicalin inhibited proliferation and promoted apoptosis by regulating the expression of MEG3/p53, indicating that baicalin may serve a role in AS by activating the MEG3/p53 signaling pathway. The present study suggested a potential mechanism underlying the protective role of baicalin in the in vitro model of AS, and these results may be used to develop novel therapeutic approaches for the affected patients.
Collapse
Affiliation(s)
- Yun Liu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Lianqun Jia
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, P.R. China
| | - Dongyu Min
- Traditional Chinese Medicine Experimental Center, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Yi Xu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Jinquan Zhu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Zengxian Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| |
Collapse
|
211
|
Vendrov AE, Sumida A, Canugovi C, Lozhkin A, Hayami T, Madamanchi NR, Runge MS. NOXA1-dependent NADPH oxidase regulates redox signaling and phenotype of vascular smooth muscle cell during atherogenesis. Redox Biol 2018; 21:101063. [PMID: 30576919 PMCID: PMC6302039 DOI: 10.1016/j.redox.2018.11.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/16/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Increased reactive oxygen species (ROS) production and inflammation are key factors in the pathogenesis of atherosclerosis. We previously reported that NOX activator 1 (NOXA1) is the critical functional homolog of p67phox for NADPH oxidase activation in mouse vascular smooth muscle cells (VSMC). Here we investigated the effects of systemic and SMC-specific deletion of Noxa1 on VSMC phenotype during atherogenesis in mice. Neointimal hyperplasia following endovascular injury was lower in Noxa1-deficient mice versus the wild-type following endovascular injury. Noxa1 deletion in Apoe-/- or Ldlr-/- mice fed a Western diet showed 50% reduction in vascular ROS and 30% reduction in aortic atherosclerotic lesion area and aortic sinus lesion volume (P < 0.01). SMC-specific deletion of Noxa1 in Apoe-/- mice (Noxa1SMC-/-/Apoe-/-) similarly decreased vascular ROS levels and atherosclerotic lesion size. TNFα-induced ROS generation, proliferation and migration were significantly attenuated in Noxa1-deficient versus wild-type VSMC. Immunofluorescence analysis of atherosclerotic lesions showed a significant decrease in cells positive for CD68 and myosin11 (22% versus 9%) and Mac3 and α-actin (17% versus 5%) in the Noxa1SMC-/-/Apoe-/- versus Apoe-/- mice. The expression of transcription factor KLF4, a modulator of VSMC phenotype, and its downstream targets – VCAM1, CCL2, and MMP2 – were significantly reduced in the lesions of Noxa1SMC-/-/Apoe-/- versus Apoe-/- mice as well as in oxidized phospholipids treated Noxa1SMC-/- versus wild-type VSMC. Our data support an important role for NOXA1-dependent NADPH oxidase activity in VSMC plasticity during restenosis and atherosclerosis, augmenting VSMC proliferation and migration and KLF4-mediated transition to macrophage-like cells, plaque inflammation, and expansion. NOXA1 is a VSMC-specific regulator of NADPH oxidase 1 activity and downstream cell signaling. NOX1 NADPH oxidase-dependent ROS generation is required for VSMC proliferation and migration after endovascular injury. NOXA1-dependent NOX1 activation of KLF4 in atherosclerotic lesions induces SMC phenotypic switch to macrophage-like cells. Atherosclerotic lesion macrophage-like cells promote plaque inflammation, matrix remodeling and increase volume expansion.
Collapse
Affiliation(s)
- Aleksandr E Vendrov
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arihiro Sumida
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chandrika Canugovi
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrey Lozhkin
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Takayuki Hayami
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R Madamanchi
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marschall S Runge
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
212
|
Coupled Modeling of Lipid Deposition, Inflammatory Response and Intraplaque Angiogenesis in Atherosclerotic Plaque. Ann Biomed Eng 2018; 47:439-452. [PMID: 30488310 DOI: 10.1007/s10439-018-02173-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
Abstract
We propose a multiphysical mathematical model by fully coupling lipid deposition, monocytes/macrophages recruitment and angiogenesis to investigate the pathophysiological responses of an atherosclerotic plaque to the dynamic changes in the microenvironment. The time evolutions of cellular (endothelial cells, macrophages, smooth muscle cells, etc.) and acellular components (low density lipoprotein, proinflammatory cytokines, extravascular plasma concentration, etc.) within the plaque microenvironment are assessed quantitatively. The thickening of the intima, the distributions of the lipid and inflammatory factors, and the intraplaque hemorrhage show a qualitative consistency with the MRI and histology data. Models with and without angiogenesis are compared to demonstrate the important role of neovasculature in the accumulation of blood-borne components in the atherosclerotic lesion by extravasation from the leaky vessel wall, leading to the formation of a lipid core and an inflammatory microenvironment, which eventually promotes plaque destabilization. This model can serve as a theoretical platform for the investigation of the pathological mechanisms of plaque progression and may contribute to the optimal design of atherosclerosis treatment strategies, such as lipid-lowering or anti-angiogenetic therapies.
Collapse
|
213
|
Atherosclerotic plaque instability in carotid arteries: miR-200c as a promising biomarker. Clin Sci (Lond) 2018; 132:2423-2436. [PMID: 30389857 DOI: 10.1042/cs20180684] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 12/19/2022]
Abstract
Early recognition of vulnerable carotid plaques could help in identifying patients at high stroke risk, who may benefit from earlier revascularisation. Nowadays, different biomarkers of plaque instability have been unravelled, among these miRNAs are promising tools for the diagnosis and treatment of atherosclerosis. Inflammation, reactive oxygen species (ROS) and endothelial dysfunction play a key role in unstable plaques genesis. We showed that miR-200c induces endothelial dysfunction, ROS production and a positive mechanism among miR-200c and miR-33a/b, two miRNAs involved in atherosclerosis progression. The goal of the present study was to determine whether miR-200c could be an atherosclerosis biomarker. Carotid plaques of patients that underwent carotid endarterectomy (CEA) were assayed for miR-200c expression. miR-200c was up-regulated in carotid plaques (n=22) and its expression was higher in unstable (n=12) compared with stable (n=10) plaques. miR-200c positively correlated with instability biomarkers (i.e. monocyte chemoattractant protein-1, cicloxigenase-2 (COX2), interleukin 6 (IL6), metalloproteinase (MMP) 1 (MMP1), 9 (MMP9)) and miR-33a/b. Moreover, miR-200c negatively correlated with stability biomarkers (i.e. zinc finger E-box binding homoeobox 1 (ZEB1), endothelial nitric oxide (NO) synthase (eNOS), forkhead boxO1 (FOXO1) and Sirtuin1 (SIRT1)) (stable plaques = 15, unstable plaques = 15). Circulating miR-200c was up-regulated before CEA in 24 patients, correlated with miR-33a/b and decreased 1 day after CEA. Interestingly, 1 month after CEA, circulating miR-200c is low in patients with stable plaques (n=11) and increased to control levels, in patients with unstable plaques (n=13). Further studies are needed to establish whether miR-200c represents a circulating biomarker of plaque instability. Our results show that miR-200c is an atherosclerotic plaque progression biomarker and suggest that it may be clinically useful to identify patients at high embolic risk.
Collapse
|
214
|
Rezvani-Sharif A, Tafazzoli-Shadpour M, Avolio A. Progressive changes of elastic moduli of arterial wall and atherosclerotic plaque components during plaque development in human coronary arteries. Med Biol Eng Comput 2018; 57:731-740. [PMID: 30374700 DOI: 10.1007/s11517-018-1910-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022]
Abstract
Stiffness of the arterial wall and atherosclerotic plaque components is a determinant of the stress field within plaques, which has been suggested to be an indicator of plaque vulnerability. The diversity and inhomogeneous structure of atherosclerotic lesions complicate the characterization of plaque components. In the present study, stiffness of the arterial wall and atherosclerotic plaque components in human coronary arteries was examined in early and developed atherosclerotic lesions. The force-spectroscopy mode of the atomic force microscope and histological examination were used for determination of elastic moduli at specified locations within samples. Fibrous cap (E = 14.1 ± 3.8 kPa) showed lower stiffness than the fibrous tissue beneath the lipid pool (E = 17.6 ± 3.2 kPa). Calcification zones (E = 96.1 ± 18.8 kPa) and lipid pools (E = 2.7 ± 1.8 kPa) were the stiffest and softest components of atherosclerotic lesions, respectively. The increase of media stiffness (%44.8) and reduction of the elastic modulus of the internal elastic lamina (%28.9) was observed in coronary arteries. Moreover, significant differences were observed between the stiffness of medial layer in diseased parts and free-plaque segments in incomplete plaques of coronary arteries. Our results can be used for better understanding of remodeling mechanisms of the arterial wall with plaque development. Graphical abstract Stiffness alteration of the arterial wall and atherosclerotic plaque components with plaque development in coronary arteries.
Collapse
Affiliation(s)
- Alireza Rezvani-Sharif
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Hafez Street, Tehran, Iran.,Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Alberto Avolio
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Hafez Street, Tehran, Iran
| |
Collapse
|
215
|
PPAR𝛾 Gene and Atherosclerosis: Genetic Polymorphisms, Epigenetics and Therapeutic Implications. Balkan J Med Genet 2018; 21:39-46. [PMID: 30425909 PMCID: PMC6231320 DOI: 10.2478/bjmg-2018-0011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is the leading cause of mortality and morbidity in the developed world. It is characterized by the formation of a plaque in the walls of middle and large arteries leading to macrovascular complications. Several risk factors are included, with diabetes being one of the most important for the onset and development of atherosclerosis. Due to an increase in the prevalence of diabetes in the world, the incidence of diabetic complications (microvascular and macrovascular) is increasing. Peroxisome proliferator-activated receptor γ (PPARγ) plays a important role in atherosclerotic processes. Peroxisome proliferator activated receptor γ belongs to the superfamily of nuclear receptors, has a great presence in fat tissue, macrophages, and regulates gene expression and most of the processes that lead to the onset and development of atherosclerosis. In this review, we discuss the basic patho-physiological mechanisms of atherosclerosis in type 2 diabetes mellitus (T2DM). Furthermore, we discuss the impact of PPARγ polymorphisms, and the epigenetic mechanisms affecting the onset of atherosclerosis, i.e, DNA methylation and demethylation, histone acetylation and deacetylation, and RNA-based mechanisms. Moreover, we add therapeutic possibilities for acting on epigenetic mechanisms in order to prevent the onset and progression of atherosclerosis.
Collapse
|
216
|
Li M, Qian M, Kyler K, Xu J. Endothelial-Vascular Smooth Muscle Cells Interactions in Atherosclerosis. Front Cardiovasc Med 2018; 5:151. [PMID: 30406116 PMCID: PMC6207093 DOI: 10.3389/fcvm.2018.00151] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory process that can eventually lead to cardiovascular disease (CVD). Despite available treatment, the prevalence of atherosclerotic CVD, which has become the leading cause of death worldwide, persists. Identification of new mechanisms of atherogenesis are highly needed in order to develop an effective therapeutic treatment. The blood vessels contain two primary major cell types: endothelial cells (EC) and vascular smooth muscle cells (VSMC). Each of these performs an essential function in sustaining vascular homeostasis. EC-VSMC communication is essential not only to development, but also to the homeostasis of mature blood vessels. Aberrant EC-VSMC interaction could promote atherogenesis. Identification of the mode of EC-VSMC crosstalk that regulates vascular functionality and sustains homeostasis may offer strategic insights for prevention and treatment of atherosclerotic CVD. Here we will review the molecular mechanisms underlying the interplay between EC and VSMC that could contribute to atherosclerosis. We also highlight open questions for future research directions.
Collapse
Affiliation(s)
- Manna Li
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Ming Qian
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Kathy Kyler
- Office of Research Administration, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| |
Collapse
|
217
|
Li C, He J, Zhong X, Gan H, Xia Y. CX3CL1/CX3CR1 Axis Contributes to Angiotensin II-Induced Vascular Smooth Muscle Cell Proliferation and Inflammatory Cytokine Production. Inflammation 2018; 41:824-834. [PMID: 29356931 DOI: 10.1007/s10753-018-0736-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Angiotensin II (Ang II) dysregulation has been determined in many diseases. The CX3CL1/CX3CR1 axis, which has a key role in cardiovascular diseases, is involved in the proliferation and inflammatory cytokine production of vascular smooth muscle cells (VSMCs). In this study, we aim to explore whether Ang II has a role in the expression of CX3CL1/CX3CR1, thus contributing to the proliferation and pro-inflammatory status of VSMCs. Cultured mouse aortic VSMCs were stimulated with 100 nmol/L of Ang II, and the expression of CX3CR1 was assessed by western blot. The results demonstrated that Ang II significantly up-regulated CX3CR1 expression in VSMCs and induced the production of reactive oxygen species (ROS) and the phosphorylation of p38 MAPK. Inhibitors of NADPH oxidase, ROS, and AT1 receptor significantly reduced Ang II-induced CX3CR1 expression. Targeted disruption of CX3CR1 by transfection with siRNA significantly attenuated Ang II-induced VSMC proliferation as well as down-regulated the expression of proliferating cell nuclear antigen (PCNA). Furthermore, CX3CR1-siRNA suppressed the effect of Ang II on stimulating Akt phosphorylation. Besides, the use of CX3CR1-siRNA decreased inflammatory cytokine production induced by Ang II treatment. Our results indicate that Ang II up-regulates CX3CR1 expression in VSMCs via NADPH oxidase/ROS/p38 MAPK pathway and that CX3CL1/CX3CR1 axis contributes to the proliferative and pro-inflammatory effects of Ang II in VSMCs.
Collapse
Affiliation(s)
- Chengsheng Li
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jin He
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaoyi Zhong
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yunfeng Xia
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
218
|
Jang SA, Park DW, Sohn EH, Lee SR, Kang SC. Hyperoside suppresses tumor necrosis factor α-mediated vascular inflammatory responses by downregulating mitogen-activated protein kinases and nuclear factor-κB signaling. Chem Biol Interact 2018; 294:48-55. [DOI: 10.1016/j.cbi.2018.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/13/2018] [Accepted: 08/17/2018] [Indexed: 01/06/2023]
|
219
|
Zhou Y, Liu X, Huang N, Chen Y. Magnesium ion leachables induce a conversion of contractile vascular smooth muscle cells to an inflammatory phenotype. J Biomed Mater Res B Appl Biomater 2018; 107:988-1001. [PMID: 30270501 DOI: 10.1002/jbm.b.34192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 05/27/2018] [Accepted: 06/12/2018] [Indexed: 12/29/2022]
Abstract
Phenotype switching is a characteristic response of vascular smooth muscle cells (vSMCs) to the dynamic microenvironment and contributes to all stages of atherosclerotic plaque. Here, we immersed pure magnesium and AZ31 alloy in the completed medium under cell culture condition, applied the resultant leaching extracts to the isolated contractile rat aortic vSMCs and investigated how vSMCs phenotypically responded to the degradation of the magnesium-based stent materials. vSMCs became more proliferative and migratory but underwent more apoptosis when exposed to the degradation products of pure magnesium; while the AZ31 extracts caused less cell division but more apoptosis, thus slowing cell moving and growing. Noticeably, both leaching extracts dramatically downregulated the contractile phenotypic genes at mRNA and protein levels while significantly induced the inflammatory adhesive molecules and cytokines. Exogenously added Mg ions excited similar transformations of vSMCs. With the liberation or supplementation of Mg2+ , the expression patterns of the pro-contractile transactivator myocardin and the pro-inflammatory transcriptional factor kruppel-like factor 4 (KLF4) were reversed. Overall, the degradation of the Mg-based materials would evoke a shift of the contractile vSMCs to an inflammatory phenotype via releasing Mg ions to induce a transition from the phenotypic control of vSMCs by the myocardin to that by the KLF4. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 988-1001, 2019.
Collapse
Affiliation(s)
- Yuehua Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xing Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Nan Huang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yuping Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacy, University of South China, Hengyang, Hunan, 421001, China.,Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
220
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Smooth muscle cell-driven vascular diseases and molecular mechanisms of VSMC plasticity. Cell Signal 2018; 52:48-64. [PMID: 30172025 DOI: 10.1016/j.cellsig.2018.08.019] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the major cell type in blood vessels. Unlike many other mature cell types in the adult body, VSMC do not terminally differentiate but retain a remarkable plasticity. Fully differentiated medial VSMCs of mature vessels maintain quiescence and express a range of genes and proteins important for contraction/dilation, which allows them to control systemic and local pressure through the regulation of vascular tone. In response to vascular injury or alterations in local environmental cues, differentiated/contractile VSMCs are capable of switching to a dedifferentiated phenotype characterized by increased proliferation, migration and extracellular matrix synthesis in concert with decreased expression of contractile markers. Imbalanced VSMC plasticity results in maladaptive phenotype alterations that ultimately lead to progression of a variety of VSMC-driven vascular diseases. The nature, extent and consequences of dysregulated VSMC phenotype alterations are diverse, reflecting the numerous environmental cues (e.g. biochemical factors, extracellular matrix components, physical) that prompt VSMC phenotype switching. In spite of decades of efforts to understand cues and processes that normally control VSMC differentiation and their disruption in VSMC-driven disease states, the crucial molecular mechanisms and signalling pathways that shape the VSMC phenotype programme have still not yet been precisely elucidated. In this article we introduce the physiological functions of vascular smooth muscle/VSMCs, outline VSMC-driven cardiovascular diseases and the concept of VSMC phenotype switching, and review molecular mechanisms that play crucial roles in the regulation of VSMC phenotypic plasticity.
Collapse
Affiliation(s)
- Agne Frismantiene
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paul Erne
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
221
|
Oberoi R, Vlacil AK, Schuett J, Schösser F, Schuett H, Tietge UJF, Schieffer B, Grote K. Anti-tumor necrosis factor-α therapy increases plaque burden in a mouse model of experimental atherosclerosis. Atherosclerosis 2018; 277:80-89. [PMID: 30176568 DOI: 10.1016/j.atherosclerosis.2018.08.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/24/2018] [Accepted: 08/24/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis is critically fueled by vascular inflammation through oxidized lipids and inflammatory cytokines such as tumor necrosis factor (TNF)-α. Genetic disruption of Tnf-α reduces atherosclerosis in experimental mouse models. However, less is known about the therapeutic potential of Tnf-α blockage by pharmacological inhibitors such as monoclonal antibodies, which are already approved for several inflammatory disorders in patients. Therefore, we investigated the effect of pharmacological TNF-α inhibition on plaque development in experimental atherosclerosis. RESULTS 10 week old male Ldlr-/- mice were divided into 4 groups (n = 7-10) and fed a high fat, high cholesterol diet for 6 and 12 weeks. Simultaneously, the mouse-specific anti-Tnf-α monoclonal antibody CNTO5048 (CNT) or a control IgG was administered. RESULTS CNT reduced circulating inflammatory markers without affecting body weight and glucose metabolism. Unexpectedly, CNT treatment increased plasma triglyceride levels and pro-atherogenic very-low-density lipoprotein (VLDL) cholesterol as well as plaque burden in the thoracoabdominal aorta and in the aortic root. In addition, we observed decreased smooth muscle cell content in the lesions and a trend towards reduced collagen deposition upon Tnf-α inhibition. Furthermore, inflammatory gene expression in the aortic arch was increased following Tnf-α inhibitor treatment. CONCLUSIONS Although up to 12-week pharmacological inhibition of TNF-α in Ldlr-/- mice diminishes systemic inflammation, experimental plaque burden and vascular inflammatory gene expression are increased, while markers of plaque stability decrease. These observations may be explained by the development of a pro-atherogenic plasma lipid profile.
Collapse
Affiliation(s)
- Raghav Oberoi
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | | | - Jutta Schuett
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | - Florian Schösser
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | - Harald Schuett
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | - Uwe J F Tietge
- Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Karsten Grote
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
222
|
Li Q, Liao C, Xu W, Li G, Hong K, Cheng X, Li J. Xeroderma Pigmentosum Group D (XPD) Inhibits the Proliferation Cycle of Vascular Smooth Muscle Cell (VSMC) by Activating Glycogen Synthase Kinase 3β (GSK3β). Med Sci Monit 2018; 24:5951-5959. [PMID: 30146633 PMCID: PMC6122044 DOI: 10.12659/msm.909614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND VSMC proliferation plays a key role in atherosclerosis, but the role of XPD in VSMC proliferation remains unknown. We investigated the expression of XPD, which is involved in cell cycle regulation, and its role in VSMC proliferation response to atherogenic stimuli. MATERIAL AND METHODS Human umbilical vein VSMCs were transfected with recombinant plasmid pEGFP-N2/XPD and pEGFP-N2 and incubated with PDGF-BB in vitro. Cell viability was determined by MTT assay. The expressions of XPD, GSK3β, p-GSK3β, CDK4, and cyclin D1 protein were detected by Western blot analysis. Cell cycle was examined by flow cytometry. RESULTS PDGF inhibited the expression of XPD in VSMCs and promoted VSMC proliferation. Overexpression of XPD significantly augmented cell cycle arrest, and attenuated protein expression levels of CDK4 and cyclin D1 in VSMCs. XPD overexpression suppressed the effects of PDGF-BB in promoting G1/S transition and accelerating protein expression levels of CDK4 and cyclin D1. XPD diminished the phosphorylation of GSK3β, and SB216763 inhibited the reduction effect of XPD on CDK4 and cyclin D1. CONCLUSIONS XPD induces VSMC cell cycle arrest, and the activation of GSK3β plays a crucial role in inhibitory effect of XPD on VSMC proliferation.
Collapse
Affiliation(s)
- Qing Li
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland).,Jiangxi Provincial Key Laboratory of Molecular Medicine, Nanchang, Jiangxi, China (mainland)
| | - Chunyao Liao
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Wang Xu
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Genlin Li
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Kui Hong
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Xiaoshu Cheng
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Juxiang Li
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
223
|
Park HJ, Kim MK, Kim Y, Bae SS, Kim HJ, Bae SK, Bae MK. Gastrin-releasing peptide promotes the migration of vascular smooth muscle cells through upregulation of matrix metalloproteinase-2 and -9. BMB Rep 2018; 50:628-633. [PMID: 29065966 PMCID: PMC5749909 DOI: 10.5483/bmbrep.2017.50.12.158] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Indexed: 12/18/2022] Open
Abstract
Gastrin-releasing peptide (GRP) has been reported to be implicated in the pathogenesis of inflammatory disorders. The migration and proliferation of vascular smooth muscle cells (VSMCs) are key components of vascular inflammation that leads to the development of atherosclerosis. The present study aimed to investigate the molecular effect of GRP on VSMC proliferation and migration. We report that GRP significantly enhanced the proliferation and migration of rat VSMCs. GRP increased mRNA and protein expression of matrix metalloproteinase-2 and -9 (MMP-2/9) in VSMCs. The induction of MMP-2/9 by GRP was regulated by the activation of the signal transducer and activator of transcription-3 (STAT3). In addition, STAT3-knockdown of VSMCs by siRNA or blockade of the GRP receptor inhibited GRP-induced migration of VSMCs. Taken together, our findings indicate that GRP promotes the migration of VSMCs through upregulation of MMP-2/9 via STAT3 activation.
Collapse
Affiliation(s)
- Hyun-Joo Park
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50610, Korea
| | - Mi-Kyoung Kim
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50610, Korea
| | - Yeon Kim
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50610, Korea
| | - Sun Sik Bae
- Department of Pharmacology, Pusan National University School of Medicine, Pusan National University, Yangsan 50610, Korea
| | - Hyung Joon Kim
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50610, Korea
| | - Soo-Kyung Bae
- Department of Dental Pharmacology, BK21 PLUS Project, School of Dentistry, Yangsan 50610, Korea
| | - Moon-Kyoung Bae
- Department of Oral Physiology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50610, Korea
| |
Collapse
|
224
|
Qiao L, Chen W. Atheroprotective effects and molecular targets of bioactive compounds from traditional Chinese medicine. Pharmacol Res 2018; 135:212-229. [PMID: 30107203 DOI: 10.1016/j.phrs.2018.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/12/2018] [Accepted: 07/12/2018] [Indexed: 01/16/2023]
Abstract
Traditional Chinese medicine (TCM) has served the Chinese people since antiquity, and is playing an important role in today's healthcare. However, there has been controversy in the use of these traditional herbs due to unclear components and absence of scientific proof. As China plans to modernize traditional medicine, successful attempts to better understand the molecular mechanisms of TCM have been made by focusing on isolating active ingredients from these remedies. In this review, we critically examined the current evidence on atheroprotective effects of bioactive compounds from TCM using in vitro or in vivo models in the past two decades. A total of 47 active compounds were included in our review, which were introduced in the order of chemical structures, source, model, efficacy and mechanism. Notablely, this review highlighted the cellular and molecular mechanisms of these active compounds in prevention and treatment of atherosclerosis. Two compounds were also involved in double-blind, randomized, placebo-controlled clinical trials (RCTs). Besides, we introduced the legislations of the People's Republic of China ensuring quality and safety of products used in TCM. In summary, studies on bioactive compounds from TCM will provide a new approach for better management of atherosclerosis.
Collapse
Affiliation(s)
- Lei Qiao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenqiang Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
225
|
Herman AB, Vrakas CN, Ray M, Kelemen SE, Sweredoski MJ, Moradian A, Haines DS, Autieri MV. FXR1 Is an IL-19-Responsive RNA-Binding Protein that Destabilizes Pro-inflammatory Transcripts in Vascular Smooth Muscle Cells. Cell Rep 2018; 24:1176-1189. [PMID: 30067974 PMCID: PMC11004729 DOI: 10.1016/j.celrep.2018.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/18/2018] [Accepted: 07/01/2018] [Indexed: 12/22/2022] Open
Abstract
This work identifies the fragile-X-related protein (FXR1) as a reciprocal regulator of HuR target transcripts in vascular smooth muscle cells (VSMCs). FXR1 was identified as an HuR-interacting protein by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The HuR-FXR1 interaction is abrogated in RNase-treated extracts, indicating that their association is tethered by mRNAs. FXR1 expression is induced in diseased but not normal arteries. siRNA knockdown of FXR1 increases the abundance and stability of inflammatory mRNAs, while overexpression of FXR1 reduces their abundance and stability. Conditioned media from FXR1 siRNA-treated VSMCs enhance activation of naive VSMCs. RNA EMSA and RIP demonstrate that FXR1 interacts with an ARE and an element in the 3' UTR of TNFα. FXR1 expression is increased in VSMCs challenged with the anti-inflammatory cytokine IL-19, and FXR1 is required for IL-19 reduction of HuR. This suggests that FXR1 is an anti-inflammation responsive, HuR counter-regulatory protein that reduces abundance of pro-inflammatory transcripts.
Collapse
Affiliation(s)
- Allison B Herman
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Christine N Vrakas
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Sheri E Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Michael J Sweredoski
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Annie Moradian
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Dale S Haines
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University Philadelphia, PA 19140, USA
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
226
|
Rezvani-Sharif A, Tafazzoli-Shadpour M, Avolio A. Mechanical Characterization of the Lamellar Structure of Human Abdominal Aorta in the Development of Atherosclerosis: An Atomic Force Microscopy Study. Cardiovasc Eng Technol 2018; 10:181-192. [PMID: 30006817 DOI: 10.1007/s13239-018-0370-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/08/2018] [Indexed: 12/16/2022]
Abstract
Atherosclerosis is a major risk factor for cardiovascular disease. However, mechanisms of interaction of atherosclerotic plaque development and local stiffness of the lamellar structure of the arterial wall are not well established. In the current study, the local Young's modulus of the wall and plaque components were determined for three different groups of healthy, mildly diseased and advanced atherosclerotic human abdominal aortas. Histological staining was performed to highlight the atherosclerotic plaque components and lamellar structure of the aortic media, consisting of concentric layers of elastin and interlamellar zones. The force spectroscopy mode of the atomic force microscopy was utilized to determine Young's moduli of aortic wall lamellae and plaque components at the micron level. The high variability of Young's moduli (E) at different locations of the atherosclerotic plaque such as the fibrous cap (E = 15.5± 2.6 kPa), calcification zone (E = 103.7±19.5 kPa), and lipid pool (E = 3.5±1.2 kPa) were observed. Reduction of elastin lamellae stiffness (18.6%), as well as stiffening of interlamellar zones (50%), were detected in the diseased portion of the medial layer of abdominal aortic wall compared to the healthy artery. Additionally, significant differences in the stiffness of both elastin lamellae and interlamellar zones were observed between the diseased wall and disease-free wall in incomplete plaques. Our results elucidate the alternation of the stiffness of different lamellae in the human abdominal aortic wall with atherosclerotic plaque development and may provide new insight on the remodeling of the aortic wall during the progression of atherosclerosis.
Collapse
Affiliation(s)
- Alireza Rezvani-Sharif
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.,Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | | | - Alberto Avolio
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
227
|
Zhai H, Qi X, Li Z, Zhang W, Li C, Ji L, Xu K, Zhong H. TIMP‑3 suppresses the proliferation and migration of SMCs from the aortic neck of atherosclerotic AAA in rabbits, via decreased MMP‑2 and MMP‑9 activity, and reduced TNF‑α expression. Mol Med Rep 2018; 18:2061-2067. [PMID: 29956789 PMCID: PMC6072177 DOI: 10.3892/mmr.2018.9224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 01/05/2017] [Indexed: 01/20/2023] Open
Abstract
The present study investigated the role of tissue inhibitor of matrix metalloproteinase‑3 (TIMP‑3) in regulating the proliferation, migration, apoptosis and activity of matrix metalloproteinase (MMP)‑2 and ‑9, during the development of an atherosclerotic abdominal artery aneurysm (AAA). Experiments were conducted using rabbit AAA neck (NA) smooth muscle cells (SMCs), to investigate the potential for TIMP‑3 to be used as a novel stent coating in preventing aortic dilation adjacent to the AAA. The atherosclerotic AAA model was induced in New Zealand white rabbits via a 6‑week high‑cholesterol diet, followed by incubation of the targeted aortic region with elastase. SMCs were isolated from the aorta adjacent to the aneurysm 30 days after AAA model induction, and stimulated with 3, 10, 30 or 100 ng/ml TIMP‑3. Cell proliferation was investigated using Cell Counting Kit‑8 reagent, migration was examined using a Boyden chamber assay and apoptotic rate was analyzed using the Annexin V‑fluorescein isothiocyanate Apoptosis Detection kit. Gelatin zymography and ELISA were used to measure the activity of MMP‑2 and MMP‑9, and the expression of tumor necrosis factor‑α (TNF‑α), respectively. Analysis of cell proliferation indicated that 10, 30 and 100 ng/ml TIMP‑3 reduced cell viability. Cell migration was decreased by 10, 30 and 100 ng/ml TIMP‑3. MMP‑2 activity was inhibited by 10, 30 and 100 ng/ml TIMP‑3, and MMP‑9 activity was suppressed by 30 and 100 ng/ml TIMP‑3. The protein levels of secreted TNF‑α were reduced by 10, 30 and 100 ng/ml TIMP‑3. The present study demonstrated the ability of 30 and 100 ng/ml TIMP‑3 to attenuate migration and proliferation, and to inhibit the activity of MMP‑2, MMP‑9 and TNF‑α secretion of NA SMCs. In conclusion, TIMP‑3 may be considered a potential therapeutic drug for use in a novel drug‑eluting stent, to attenuate the progressive dilation of the aortic NA.
Collapse
Affiliation(s)
- Huan Zhai
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xun Qi
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zixuan Li
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Wei Zhang
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chenguang Li
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lu Ji
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ke Xu
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hongshan Zhong
- Department of Interventional Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
228
|
Zhou B, Prim DA, Romito EJ, McNamara LP, Spinale FG, Shazly T, Eberth JF. Contractile Smooth Muscle and Active Stress Generation in Porcine Common Carotids. J Biomech Eng 2018; 140:2654977. [PMID: 28975258 DOI: 10.1115/1.4037949] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Indexed: 01/22/2023]
Abstract
The mechanical response of intact blood vessels to applied loads can be delineated into passive and active components using an isometric decomposition approach. Whereas the passive response is due predominantly to the extracellular matrix (ECM) proteins and amorphous ground substance, the active response depends on the presence of smooth muscle cells (SMCs) and the contractile machinery activated within those cells. To better understand determinants of active stress generation within the vascular wall, we subjected porcine common carotid arteries (CCAs) to biaxial inflation-extension testing under maximally contracted or passive SMC conditions and semiquantitatively measured two known markers of the contractile SMC phenotype: smoothelin and smooth muscle-myosin heavy chain (SM-MHC). Using isometric decomposition and established constitutive models, an intuitive but novel correlation between the magnitude of active stress generation and the relative abundance of smoothelin and SM-MHC emerged. Our results reiterate the importance of stretch-dependent active stress generation to the total mechanical response. Overall these findings can be used to decouple the mechanical contribution of SMCs from the ECM and is therefore a powerful tool in the analysis of disease states and potential therapies where both constituent are altered.
Collapse
Affiliation(s)
- Boran Zhou
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - David A Prim
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208
| | - Eva J Romito
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208; Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC 29208
| | - Liam P McNamara
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC 29208; School of Medicine, Department of Cell Biology and Anatomy, University of South Carolina, Columbia, SC 29208
| | - Tarek Shazly
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208; College of Engineering and Computing, Department of Mechanical Engineering, University of South Carolina, Columbia, SC 29208
| | - John F Eberth
- College of Engineering and Computing, Biomedical Engineering Program, University of South Carolina, Columbia, SC 29208; School of Medicine, Department of Cell Biology and Anatomy, University of South Carolina, Columbia, SC 29208 e-mail:
| |
Collapse
|
229
|
Kimura S, Noguchi H, Nanbu U, Wang KY, Sasaguri Y, Nakayama T. Relationship between CCL22 Expression by Vascular Smooth Muscle Cells and Macrophage Histamine Receptors in Atherosclerosis. J Atheroscler Thromb 2018; 25:1240-1254. [PMID: 29794410 PMCID: PMC6249366 DOI: 10.5551/jat.44297] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIM CCL22, mainly synthesized by monocyte-derived alternative (M2) macrophages, belongs to the CC family of chemokines and is involved in monocyte migration and recruitment. We have previously investigated CCL22 and histamine in atherosclerosis. Here, we investigated the hypothesis that CCL22 is involved in atherosclerosis, which is influenced by the differentiation of macrophage phenotypes via histamine. METHODS CCL22 expression was investigated in human carotid arteries and coronary arteries with bare metal stents. Ligated carotid arteries of wild-type (C57BL/6J) and apolipoprotein E-deficient mice were also used as atherosclerotic models. The localization and expression of CCL22 and classical (M1)-like and M2-like macrophages in various human and mouse atherosclerotic lesions were investigated by immunohistochemical examination and quantitative real-time polymerase chain reaction. Histamine is expressed in atherosclerosis, and it induces inflammation and immunity. Human- and mice-derived monocytes and macrophages were used to examine the role of histamine in macrophage differentiation and CCL22-expression. Macrophages derived from histamine receptor 1 (H1R)- and 2 (H2R)-knockout (KO) mice were also examined. RESULTS Atherosclerotic lesions showed a distribution of heterogeneous macrophage phenotypes with M1-like and M2-like macrophage dominant sites. CCL22 was distributed in sparse areas of vascular smooth muscle cells (VSMCs) and associated with M2-like macrophages. Moreover, H2R stimulation was associated with CCL22 expression via M2-like macrophage dominant differentiation. CONCLUSION The expression of M1- or M2-like macrophages in atherosclerosis were observed to be dependent on the distribution of VSMCs owing to differences in causal stimuli and the switching of histamine receptors via Th1 or Th2 cytokines. These results suggest that CCL22 may control atherosclerosis.
Collapse
Affiliation(s)
- Satoshi Kimura
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health
| | - Hirotsugu Noguchi
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health
| | - Uki Nanbu
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health
| | - Ke-Yong Wang
- Shared-Use Research Center, University of Occupational and Environmental Health
| | - Yasuyuki Sasaguri
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health
| | - Toshiyuki Nakayama
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health
| |
Collapse
|
230
|
Wang W, Chen Y, Bai L, Zhao S, Wang R, Liu B, Zhang Y, Fan J, Liu E. Transcriptomic analysis of the liver of cholesterol-fed rabbits reveals altered hepatic lipid metabolism and inflammatory response. Sci Rep 2018; 8:6437. [PMID: 29692426 PMCID: PMC5915436 DOI: 10.1038/s41598-018-24813-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 04/10/2018] [Indexed: 12/11/2022] Open
Abstract
Rabbits are a suitable animal model for atherosclerosis due to their sensitivity to dietary cholesterol. Moreover, rabbits have lipoprotein profiles that are more similar to humans than those of other laboratory animals. However, little is known about the transcriptomic information related to atherosclerosis in rabbits. We aimed to determine the changes in the livers of rabbits fed a normal chow diet (control) or high cholesterol diet (HCD) by histological examinations and RNA sequencing analysis. Compared with the control group, the lipid levels and small LDL subfractions in plasma were increased, and aortic atherosclerotic plaques were formed in the HCD group. Most importantly, HCD resulted in lipid accumulation and inflammation in the livers. Transcriptomic analysis of the liver showed that HCD induces 1183 differentially expressed genes (DEGs) that mainly participate in the regulation of inflammation and lipid metabolism. Furthermore, the signaling pathways involved in inflammation and lipid metabolism were enriched by KEGG pathway analysis. In addition, hepatic DEGs of the HCD group were further validated by real-time PCR. These results suggest that HCD causes liver lipid accumulation and inflammatory response. Although the relationships between these hepatic changes and atherogenesis need further investigation, these findings provide a fundamental framework for future research on human atherosclerosis using rabbit models.
Collapse
Affiliation(s)
- Weirong Wang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yulong Chen
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Liang Bai
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Sihai Zhao
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Rong Wang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Baoning Liu
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yali Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, 710061, China.
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
231
|
Zhang Y, Qian X, Sun X, Lin C, Jing Y, Yao Y, Ma Z, Kuai M, Lu Y, Kong X, Chen Q, Wu X, Zhao X, Li Y, Bian H. Liuwei Dihuang, a traditional Chinese medicinal formula, inhibits proliferation and migration of vascular smooth muscle cells via modulation of estrogen receptors. Int J Mol Med 2018; 42:31-40. [PMID: 29693116 PMCID: PMC5979928 DOI: 10.3892/ijmm.2018.3622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 11/10/2017] [Indexed: 01/27/2023] Open
Abstract
The phenotypic modulation of vascular smooth muscle cells (VSMCs) serves an important role in atherosclerosis-induced vascular alterations, including vascular remodeling. However, the precise mechanisms underlying VSMC phenotypic modulation remain to be elucidated. Our previous study demonstrated that Liuwei Dihuang formula (LWDHF) could improve menopausal atherosclerosis by upregulating the expression of estrogen receptors (ERs). The present study examined the role of ERs in the effects of LWDHF on VSMC phenotypic modulation. VSMC proliferation and cell cycle progression were examined by MTT assay and flow cytometry, respectively. The expression levels of α-smooth muscle actin, osteopontin and ERs were determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. Cell ultrastructure was observed under an electron microscope. F-actin polymerization was detected by fluorescein isothiocyanate-phalloidin staining using fluorescence microscopy. A modified Boyden chamber assay was employed to assess VSMCs migration. Small interfering (si)RNA technology was used to examine the role of ERα in the effects of LWDHF on phenotypic modulation. The results indicated that LWDHF (3-12 µg/ml) inhibited proliferation and induced a cell cycle arrest in VSMCs treated with angiotensin II (Ang II; 100 nM) in a concentration-dependent manner. In addition, Ang II-stimulated migration of VSMCs and reorganization of actin were markedly inhibited by treatment with 12 µg/ml LWDHF. Results of RT-qPCR and western blotting demonstrated that LWDHF markedly stimulated transcription and expression of ERα and ERβ, and inhibited VSMC synthetic phenotype. Furthermore, LWDHF-induced inhibition of phenotypic switching was partially suppressed by tamoxifen, and transfection with ERα siRNA markedly abolished the effects of LWDHF on VSMC phenotypic switching. In conclusion, these results revealed that ERα served an important role in LWDHF-induced regulation of the VSMC phenotype, including proliferation and migration.
Collapse
Affiliation(s)
- Yayun Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xing Qian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xin Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Chao Lin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yi Jing
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yuan Yao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Zhi Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Meiyu Kuai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Ying Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xueyun Kong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Qi Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xiang Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xuan Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yu Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Huimin Bian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
232
|
Liu Y, Peng W, Qu K, Lin X, Zeng Z, Chen J, Wei D, Wang Z. TET2: A Novel Epigenetic Regulator and Potential Intervention Target for Atherosclerosis. DNA Cell Biol 2018; 37:517-523. [PMID: 29653065 DOI: 10.1089/dna.2017.4118] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Atherosclerosis is the underlying cause of cardio-cerebrovascular disease. However, the mechanisms of atherosclerosis are still unclear. The modification of DNA methylation has an important role in atherosclerosis development. As a member of the Ten-eleven translocation (TET) family, TET methylcytosine dioxygenase 2 (TET2) can modify DNA methylation by catalyzing 5-methylcytosine to 5-hydroxymethylcytosine and mediate DNA demethylation. Recent findings suggest that TET2 is related to the phenotype transformation of vascular smooth muscle cells, endothelial dysfunction, and inflammation of macrophage, the key factors of atherosclerosis. Therefore, TET2 may be a potential target for atherosclerosis treatment. This review will elaborate the recent findings that suggest the role of TET2 in atherosclerosis.
Collapse
Affiliation(s)
- Yami Liu
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Wen Peng
- 2 Department of Spine Surgery, The First Affiliated Hospital, University of South China , Hengyang, China
| | - Kai Qu
- 3 College of Bioengineering, Chongqing University , Chongqing, China
| | - Xiaolong Lin
- 4 Department of Pathology, The Third People's Hospital of Huizhou , Huizhou, China
| | - Zhaolin Zeng
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Jiaojiao Chen
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Dangheng Wei
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| | - Zuo Wang
- 1 Key Laboratory for Atherosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China , Hengyang, China
| |
Collapse
|
233
|
Autieri MV. IL-19 and Other IL-20 Family Member Cytokines in Vascular Inflammatory Diseases. Front Immunol 2018; 9:700. [PMID: 29681905 PMCID: PMC5897441 DOI: 10.3389/fimmu.2018.00700] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/21/2018] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease remains a major medical and socioeconomic burden in developed and developing countries and will increase with an aging and increasingly sedentary society. Many vascular diseases and atherosclerotic vascular disease, in particular, are essentially inflammatory disorders, involving multiple cell types. Communication between these cells is initiated and sustained by a complex network of cytokines and their receptors. The interleukin (IL)-20 family members, IL-19, IL-20, IL-22, and IL-24, initiate, sustain, and drive the progression of vascular disease. They are important in vascular disease as they facilitate a bidirectional cross-talk between resident vascular cells with immune cells. These cytokines are grouped into the same family based on shared common receptor subunits and signaling pathways. This communication is varied and can result in exacerbation, attenuation, and even repair of the vasculature. We will briefly review what is known about IL-20, IL-22, and IL-24 in cardiovascular biology. Because IL-19 is the most studied member of this family in terms of its role in vascular pathophysiological processes, the major emphasis of this review will focus on the expression and atheroprotective roles of IL-19 in vascular inflammatory disease.
Collapse
Affiliation(s)
- Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
234
|
Li JY, Liu CP, Shiao WC, Jayakumar T, Li YS, Chang NC, Huang SY, Hsieh CY. Inhibitory effect of PDGF-BB and serum-stimulated responses in vascular smooth muscle cell proliferation by hinokitiol via up-regulation of p21 and p53. Arch Med Sci 2018; 14:579-587. [PMID: 29765446 PMCID: PMC5949921 DOI: 10.5114/aoms.2018.75085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/30/2015] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Vascular smooth muscle cell (VSMC) proliferation plays a major role in the progression of vascular diseases. In the present study, we established the efficacy and the mechanisms of action of hinokitiol, a tropolone derivative found in Chamaecyparis taiwanensis, Cupressaceae, in relation to platelet-derived growth factor-BB (PDGF-BB) and serum-dependent VSMC proliferation. MATERIAL AND METHODS Primary cultured rat VSMCs were pre-treated with hinokitiol and then stimulated by PDGF-BB (10 ng/ml) or serum (10% fetal bovine serum). Cell proliferation and cytotoxicity were determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and lactose dehydrogenase assay, respectively. The degree of DNA synthesis was evaluated by BrdU-incorporation measurements and observed using confocal microscopy. Immunoblotting was utilized to determine the protein level of p-extracellular signal-regulated kinase (ERK) 1/2, p-Akt, p-phosphoinositide 3-kinase (PI3K), p-Janus kinase 2 (JAK2), p-p53, and p21Cip1. The promoter activity of p21 and p53 activity were measured by dual luciferase reporter assay. RESULTS Treatment with hinokitiol (1-10 μM) inhibited PDGF-BB and serum-induced VSMC proliferation and DNA synthesis in a concentration-dependent manner. Cytotoxicity was not observed in hinokitiol-treated VSMCs at the studied concentrations. Pre-incubation of VSMCs with hinokitiol did not alter PDGF-BB-induced phosphorylation of ERK1/2, Akt, PI3K or JAK2. Interestingly, hinokitiol induced promoter activity of p21 and p21 protein expression in VSMCs. Furthermore, hinokitiol augmented p53 protein phosphorylation and subsequently led to enhanced p53 activity. CONCLUSIONS These data suggest that the anti-proliferative effects of hinokitiol in VSMCs may be mediated by activation of p21 and p53 signaling pathways, and it may contribute to the prevention of vascular diseases associated with VSMC proliferation.
Collapse
Affiliation(s)
- Jiun-Yi Li
- Department of Cardiovascular Surgery, Mackay Memorial Hospital, and Mackay Medical College, Taipei, Taiwan
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Ping Liu
- Department of Cardiology, Yuan’s General Hospital, Kaohsiung, Taiwan
| | - Wei-Cheng Shiao
- Department of Internal Medicine, Yuan’s General Hospital, Kaohsiung, Taiwan
| | - Thanasekaran Jayakumar
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Shin Li
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nen-Chung Chang
- Department of Cardiology, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ying Hsieh
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
235
|
MicroRNA-145 alleviates high glucose-induced proliferation and migration of vascular smooth muscle cells through targeting ROCK1. Biomed Pharmacother 2018; 99:81-86. [DOI: 10.1016/j.biopha.2018.01.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/20/2017] [Accepted: 01/03/2018] [Indexed: 11/22/2022] Open
|
236
|
Fu Y, Chang Y, Chen S, Li Y, Chen Y, Sun G, Yu S, Ye N, Li C, Sun Y. BAG3 promotes the phenotypic transformation of primary rat vascular smooth muscle cells via TRAIL. Int J Mol Med 2018; 41:2917-2926. [PMID: 29484366 DOI: 10.3892/ijmm.2018.3493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/08/2018] [Indexed: 11/05/2022] Open
Abstract
Under normal physiological condition, the mature vascular smooth muscle cells (VSMCs) show differentiated phenotype. In response to various environmental stimuluses, VSMCs convert from the differentiated phenotype to dedifferentiated phenotype characterized by the increased ability of proliferation/migration and the reduction of contractile ability. The phenotypic transformation of VSMCs played an important role in atherosclerosis. Both Bcl-2-associated athanogene 3 (BAG3) and tumor necrosis factor-related apopt-osis inducing ligand (TRAIL) involved in apoptosis. The relationship between BAG3 and TRAIL and their effects the proliferation and migration in VSMCs are rarely reported. This study investigated the effects of BAG3 on the phenotypic modulation and the potential underlying mechanisms in primary rat VSMCs. Primary rat VSMCs were extracted and cultured in vitro. Cell proliferation was detected by cell counting, real-time cell analyzer (RTCA) and EdU incorporation. Cell migration was detected by wound healing, Transwell and RTCA. BAG3 and TRAIL were detected using real-time PCR and western blotting and the secreted proteins in the cultured media by dot blot. The expression of BAG3 increased with continued passages in cultured primary VSMCs. BAG3 promoted the proliferation and migration of primary rat VSMC in a time-dependent manner. BAG3 significantly increased the expression of TRAIL while had no effects on its receptors. TRAIL knockdown or blocking by neutralizing antibody inhibited the proliferation of VSMCs induced by BAG3. TRAIL knockdown exerted no obvious influence on the migration of VSMCs. Based on this study, we report for the first time that BAG3 was expressed in cultured primary rat VSMCs and the expression of BAG3 increased with continued passages. Furthermore, BAG3 promoted the proliferation of VSMCs via increasing the expression of TRAIL. In addition, we also demonstrated that BAG3 promoted the migration of VSMCs independent of TRAIL upregulation.
Collapse
Affiliation(s)
- Yao Fu
- Department of Cardiology, Shenjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ye Chang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shuang Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuan Li
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yintao Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shasha Yu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ning Ye
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chao Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
237
|
Pai VC, Lo IC, Huang YW, Tsai IC, Cheng HP, Shi GY, Wu HL, Jiang MJ. The chondroitin sulfate moiety mediates thrombomodulin-enhanced adhesion and migration of vascular smooth muscle cells. J Biomed Sci 2018; 25:14. [PMID: 29439742 PMCID: PMC5809974 DOI: 10.1186/s12929-018-0415-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/30/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Thrombomodulin (TM), a transmembrane glycoprotein highly expressed in endothelial cells (ECs), is a potent anticoagulant maintaining circulation homeostasis. Under inflammatory states, TM expression is drastically reduced in ECs while vascular smooth muscle cells (VSMCs) show a robust expression of TM. The functional role of TM in VSMCs remains elusive. METHODS We examined the role of TM in VSMCs activities in human aortic VSMCs stimulated with platelet-derived growth factor-BB (PDGF-BB). Using rat embryonic aorta-derived A7r5 VSMCs which do not express TM, the role of the chondroitin sulfate (CS) moiety of TM in VSMCs was delineated with cells expressing wild-type TM and the CS-devoid TM mutant. RESULTS Expression of TM enhanced cell migration and adhesion/spreading onto type I collagen, but had no effect on cell proliferation. Knocking down TM with short hairpin RNA reduced PDGF-stimulated adhesion and migration of human aortic VSMCs. In A7r5 cells, TM-mediated cell adhesion was eradicated by pretreatment with chondroitinase ABC which degrades CS moiety. Furthermore, the TM mutant (TMS490, 492A) devoid of CS moiety failed to increase cell adhesion, spreading or migration. Wild-type TM, but not TMS490, 492A, increased focal adhesion kinase (FAK) activation during cell adhesion, and TM-enhanced cell migration was abolished by a function-blocking anti-integrin β1 antibody. CONCLUSION Chondroitin sulfate modification is required for TM-mediated activation of β1-integrin and FAK, thereby enhancing adhesion and migration activity of VSMCs.
Collapse
Affiliation(s)
- Vincent Chunpeng Pai
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - I-Chung Lo
- Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yan Wun Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - I-Ching Tsai
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - Hui-Pin Cheng
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan
| | - Guey-Yueh Shi
- Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Hua-Lin Wu
- Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Meei Jyh Jiang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, 1 Ta-Hsueh Road, Tainan, 70101, Taiwan. .,Cardiovascular Research Center, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
238
|
Celeng C, de Keizer B, Merkely B, de Jong P, Leiner T, Takx RAP. PET Molecular Targets and Near-Infrared Fluorescence Imaging of Atherosclerosis. Curr Cardiol Rep 2018; 20:11. [PMID: 29435774 PMCID: PMC5809554 DOI: 10.1007/s11886-018-0953-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW With this review, we aim to summarize the role of positron emission tomography (PET) and near-infrared fluorescence imaging (NIRF) in the detection of atherosclerosis. RECENT FINDINGS 18F-FDG is an established measure of increased macrophage activity. However, due to its low specificity, new radiotracers have emerged for more specific detection of vascular inflammation and other high-risk plaque features such as microcalcification and neovascularization. Novel NIRF probes are engineered to sense endothelial damage as an early sign of plaque erosion as well as oxidized low-density lipoprotein (oxLDL) as a prime target for atherosclerosis. Integrated NIRF/OCT (optical coherence tomography) catheters enable to detect stent-associated microthrombi. Novel radiotracers can improve specificity of PET for imaging atherosclerosis. Advanced NIRF probes show promise for future application in human. Intravascular NIRF might play a prominent role in the detection of stent-induced vascular injury.
Collapse
Affiliation(s)
- Csilla Celeng
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - Bart de Keizer
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Gaál József street 9, Budapest, 1122, Hungary
| | - Pim de Jong
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Tim Leiner
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Richard A P Takx
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|
239
|
Przybylska D, Janiszewska D, Goździk A, Bielak-Zmijewska A, Sunderland P, Sikora E, Mosieniak G. NOX4 downregulation leads to senescence of human vascular smooth muscle cells. Oncotarget 2018; 7:66429-66443. [PMID: 27655718 PMCID: PMC5341811 DOI: 10.18632/oncotarget.12079] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/12/2016] [Indexed: 11/25/2022] Open
Abstract
Senescence is a stress response characterized by an irreversible growth arrest and alterations in certain cell functions. It is believed that both double-strand DNA breaks (DSB) and increased ROS level are the main culprit of senescence. Excessive ROS production is also particularly important in the development of a number of cardiovascular disorders. In this context the involvement of professional ROS-producing enzymes, NADPH oxidases (NOX), was postulated. In contrary to the common knowledge, we have shown that not only increased ROS production but also diminished ROS level could be involved in the induction of senescence. Accordingly, our studies revealed that stress-induced premature senescence (SIPS) of vascular smooth muscle cells (VSMCs) induced by doxorubicin or H2O2, correlates with increased level of DSB and ROS. On the other hand, both SIPS and replicative senescence were accompanied by diminished expression of NOX4. Moreover, inhibition of NOX activity or decrease of NOX4 expression led to permanent growth arrest of VSMCs and secretion of interleukins and VEGF. Interestingly, cells undergoing senescence due to NOX4 depletion neither acquired DSB nor activated DNA damage response. Instead, transient induction of the p27, upregulation of HIF-1alpha, decreased expression of cyclin D1 and hypophosphorylated Rb was observed. Our results showed that lowering the level of ROS-producing enzyme - NOX4 oxidase below physiological level leads to cellular senescence of VSMCs which is correlated with secretion of pro-inflammatory cytokines. Thus the use of specific NOX4 inhibitors for pharmacotherapy of vascular diseases should be carefully considered.
Collapse
Affiliation(s)
- Dorota Przybylska
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Dorota Janiszewska
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Aleksandra Goździk
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Anna Bielak-Zmijewska
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Sunderland
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Grażyna Mosieniak
- Laboratory of Molecular Bases of Aging, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
240
|
H19 knockdown suppresses proliferation and induces apoptosis by regulating miR-148b/WNT/β-catenin in ox-LDL -stimulated vascular smooth muscle cells. J Biomed Sci 2018; 25:11. [PMID: 29415742 PMCID: PMC5804091 DOI: 10.1186/s12929-018-0418-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/02/2018] [Indexed: 02/08/2023] Open
Abstract
Background Long non-coding RNAs (lncRNAs) have been identified as critical regulators in the development of atherosclerosis (AS). Here, we focused on discussing roles and molecular mechanisms of lncRNA H19 in vascular smooth muscle cells (VSMCs) progression. Methods RT-qPCR assay was used to detect the expression patterns of H19 and miR-148b in clinical samples and cells. Cell proliferative ability was evaluated by CCK-8 and colony formation assays. Cell apoptotic capacity was assessed by apoptotic cell percentage and the caspase-3 activity. Bioinformatics analysis, luciferase and RNA immunoprecipitation (RIP) assays were employed to demonstrate cell percentage and the relationship among H19, miR-148b and wnt family member 1 (WNT1). Western blot assay was performed to determine expressions of proliferating cell nuclear antigen (PCNA), ki-67, Bax, Bcl-2, WNT1, β-catenin, C-myc and E-cadherin. Results The level of H19 was increased and miR-148b expression was decreased in human AS patient serums and oxidized low-density lipoprotein (ox-LDL)-stimulated human aorta vascular smooth muscle cells (HA-VSMCs). H19 knockdown suppressed proliferation and promoted apoptosis in HA-VSMCs following the treatment of ox-LDL. H19 inhibited miR-148b expression by direct interaction. Moreover, miR-148b inhibitor could reverse the effects of H19 depletion on proliferation and apoptosis in ox-LDL-stimulated HA-VSMCs. Further mechanical explorations showed that WNT1 was a target of miR-148b and H19 acted as a competing endogenous RNA (ceRNA) of miR-148b to enhance WNT1 expression. Furthermore, miR-148 inhibitor exerted its pro-proliferation and anti-apoptosis effects through activating WNT/β-catenin signaling in ox-LDL-stimulated HA-VSMCs. Conclusion H19 facilitated proliferation and inhibited apoptosis through modulating WNT/β-catenin signaling pathway via miR-148b in ox-LDL-stimulated HA-VSMCs, implicating the potential values of H19 in AS therapy.
Collapse
|
241
|
Goikuria H, Vandenbroeck K, Alloza I. Inflammation in human carotid atheroma plaques. Cytokine Growth Factor Rev 2018; 39:62-70. [PMID: 29396056 DOI: 10.1016/j.cytogfr.2018.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 12/31/2022]
Abstract
Inflammation in carotid atherosclerotic plaque is linked to plaque rupture and cerebrovascular accidents. The balance between pro- and anti-inflammatory mediators governs development of the plaque, and may mediate enhancement of lesion broadening or, on the contrary, delay progression. In addition to macrophages and endothelial cells, smooth muscle cells (SMCs), which are the dominant cell subset in advanced plaques, are crucial players in carotid atherosclerosis development given their ability to differentiate into distinct phenotypes in reponse to specific signals received from the environment of the lesion. Carotid atheroma SMCs actively contribute to the inflammation in the lesion because of their acquired capacity to produce inflammatory mediators. We review the successive stages of carotid atheroma plaque formation via fatty streak early-stage toward more advanced rupture-prone lesions and document involvement of cytokines and chemokines and their cellular sources and targets in plaque progression and rupture.
Collapse
Affiliation(s)
- Haize Goikuria
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain
| | - Koen Vandenbroeck
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Iraide Alloza
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain.
| |
Collapse
|
242
|
Wang Y, Wang Y, Li GR. TRPC1/TRPC3 channels mediate lysophosphatidylcholine-induced apoptosis in cultured human coronary artery smooth muscles cells. Oncotarget 2018; 7:50937-50951. [PMID: 27472391 PMCID: PMC5239449 DOI: 10.18632/oncotarget.10853] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/09/2016] [Indexed: 02/05/2023] Open
Abstract
The earlier study showed that lysophosphatidylcholine (lysoPC) induced apoptosis in human coronary artery smooth muscle cells (SMCs); however, the related molecular mechanisms are not fully understood. The present study investigated how lysoPC induces apoptosis in cultured human coronary artery SMCs using cell viability assay, flow cytometry, confocal microscopy, and molecular biological approaches. We found that lysoPC reduced cell viability in human coronary artery SMCs by eliciting a remarkable Ca2+ influx. The effect was antagonized by La3+, SKF-96365, or Pyr3 as well as by silencing TRPC1 or TRPC3. Co-immunoprecipitation revealed that TRPC1 and TRPC3 had protein-protein interaction. Silencing TRPC1 or TRPC3 countered the lysoPC-induced increase of Ca2+ influx and apoptosis, and the pro-apoptotic proteins Bax and cleaved caspase-3 and decrease of the anti-apoptotic protein Bcl-2 and the survival kinase pAkt. These results demonstrate the novel information that TRPC1/TRPC3 channels mediate lysoPC-induced Ca2+ influx and apoptosis via activating the pro-apoptotic proteins Bax and cleaved caspase-3 and inhibiting the anti-apoptotic protein Bcl-2 and the survival kinase pAkt in human coronary artery SMCs, which implies that TRPC1/TRC3 channels may be the therapeutic target of lysoPC-induced disorders such as atherosclerosis.
Collapse
Affiliation(s)
- Yuan Wang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Medical School of Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
243
|
Lu X, Yin D, Zhou B, Li T. MiR-135a Promotes Inflammatory Responses of Vascular Smooth Muscle Cells From db/db Mice via Downregulation of FOXO1. Int Heart J 2018; 59:170-179. [PMID: 29332916 DOI: 10.1536/ihj.17-040] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
It has been shown that microRNAs (miRNAs) greatly affect the functions of vascular smooth muscle cells (VSMC), but the effects of mRNAs under diabetic conditions remain unclear.Using a model of diabetic db/db mice, we studied the functions of microRNA-135a (miR-135a) during VSMC dysfunction.Compared to control WT mice, miR-135a expression in VSMC was significantly increased while the level of forkhead box O1 (FOXO1) protein decreased significantly. After transfecting miR-135a mimics into VSMC, the expression of FOXO1 was decreased, while cyclooxygenase-2 (COX-2) and monocyte chemoattractant protein-1 (MCP-1) expression levels were increased, thus promoting the interaction between monocytes and WT VSMC. On the other hand, transfection of an miR-135a inhibitor reversed the activated interaction between monocytes and db/db VSMC. The pro-inflammatory responses could also be enhanced by using siRNAs to silence the FOXO1 gene in WT VSMC, suggesting a negative regulatory role of FOXO1. FOXO1 siRNAs and miR-135a mimics could both enhance the transcriptional activity of COX-2 promoter. Using chromatin immunoprecipitation, we found that in db/db VSMC, the occupancy in promoter regions of inflammatory genes by FOXO1 was reduced.miR-135a increased the inflammatory responses of VSMC involved in complications of vascular diseases by downregulating the expression of FOXO1.
Collapse
Affiliation(s)
- Xiaochun Lu
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital
| | - Dawei Yin
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital
| | - Bo Zhou
- Department of Geriatrics, the Affiliated Zhongda Hospital of Southeast University
| | - Tieling Li
- Department of Cadre Clinic, Chinese People's Liberation Army General Hospital
| |
Collapse
|
244
|
Zhang L, Shao J, Zhou Y, Chen H, Qi H, Wang Y, Chen L, Zhu Y, Zhang M, Chen L, Du Y, Zhong M, Shi X, Li Q. Inhibition of PDGF-BB-induced proliferation and migration in VSMCs by proanthocyanidin A2: Involvement of KDR and Jak-2/STAT-3/cPLA 2 signaling pathways. Biomed Pharmacother 2018; 98:847-855. [PMID: 29571255 DOI: 10.1016/j.biopha.2018.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/29/2017] [Accepted: 01/03/2018] [Indexed: 12/21/2022] Open
Abstract
Proanthocyanidin A2 (PA2), one of A-type proanthocyanidins, has been shown to harbor a broad spectrum of pharmacological activities, including anti-inflammatory, antioxidant, anti-HIV, anti-CDV and anti-?-glucosidase activities. However, little is known about the role for PA2 in regulating PDGF-induced VSMC proliferation and migration. In the present study, we investigated the possible effects of PA2 on PDGF-BB-induced proliferation, migration and inflammation in VSMCs in vitro to mimic a postangioplasty PDGF shedding condition. Herein, the data clearly show that PA2 markedly inhibited proliferation, migration and inflammatory responses at 0-30??g/ml concentration in VSMCs in vitro. 10-30??g/ml PA2 inhibited PDGF-mediated NAD(P)H oxidase activation and intracellular ROS formation in VSMCs. Furthermore, the effects exerted by PA2 involve the participation of KDR and Jak-2/STAT-3/cPLA2 signaling pathways. These data also highlight the possible therapeutic use of PA2 in vascular proliferative diseases, where abnormal proliferation and migration play important pathological roles.
Collapse
Affiliation(s)
- Liudi Zhang
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Jie Shao
- Department of General Surgery, Huashan Hospital North, Shanghai 201907, China
| | - Yufu Zhou
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Haifei Chen
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Yi Wang
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Lu Chen
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Yongjun Zhu
- Department of Cardio-thoracic surgery, Huashan Hospital, Shanghai 200040, China.
| | - Meng Zhang
- Brunswick Laboratories (China), Suzhou Industrial Park 215021, China
| | - Li Chen
- Pharmacy Department, Xuhui district Central Hospital, 966 Huai Hai M Road, Shanghai 200031, China
| | - Yongli Du
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan 250353, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Xiaojin Shi
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital North, Shanghai 201907, China.
| |
Collapse
|
245
|
Yang H, Zhao C, Li R, Shen C, Cai X, Sun L, Luo C, Yin Y. Noninvasive and prospective diagnosis of coronary heart disease with urine using surface-enhanced Raman spectroscopy. Analyst 2018; 143:2235-2242. [DOI: 10.1039/c7an02022h] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The SERS spectra of human urine could provide valuable information for the noninvasive and prospective diagnosis of patients with suspected CHD.
Collapse
Affiliation(s)
- Huinan Yang
- School of Energy and Power Engineering
- University of Shanghai for Science and Technology
- 200093 Shanghai
- China
| | - Chang Zhao
- School of Energy and Power Engineering
- University of Shanghai for Science and Technology
- 200093 Shanghai
- China
| | - Rong Li
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine
- 200093 Shanghai
- China
| | - Chengxing Shen
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine
- 200093 Shanghai
- China
| | - Xiaoshu Cai
- School of Energy and Power Engineering
- University of Shanghai for Science and Technology
- 200093 Shanghai
- China
| | - Li Sun
- School of Energy and Power Engineering
- University of Shanghai for Science and Technology
- 200093 Shanghai
- China
| | - Chengfang Luo
- School of Energy and Power Engineering
- University of Shanghai for Science and Technology
- 200093 Shanghai
- China
| | - Yuechao Yin
- School of Energy and Power Engineering
- University of Shanghai for Science and Technology
- 200093 Shanghai
- China
| |
Collapse
|
246
|
Inflammation-regulated mRNA stability and the progression of vascular inflammatory diseases. Clin Sci (Lond) 2017; 131:2687-2699. [PMID: 29109302 DOI: 10.1042/cs20171373] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease remains a major medical and socioeconomic burden in developed and developing societies, and will increase with an aging and increasingly sedentary society. Vascular disease and atherosclerotic vascular syndromes are essentially inflammatory disorders, and transcriptional and post-transcriptional processes play essential roles in the ability of resident vascular and inflammatory cells to adapt to environmental stimuli. The regulation of mRNA translocation, stability, and translation are key processes of post-transcriptional regulation that permit these cells to rapidly respond to inflammatory stimuli. For the most part, these processes are controlled by elements in the 3'-UTR of labile, proinflammatory transcripts. Since proinflammatory transcripts almost exclusively contain AU-rich elements (AREs), this represents a tightly regulated and specific mechanism for initiation and maintenance of the proinflammatory phenotype. RNA-binding proteins (RBPs) recognize cis elements in 3'-UTR, and regulate each of these processes, but there is little literature exploring the concept that RBPs themselves can be directly regulated by inflammatory stimuli. Conceptually, inflammation-responsive RBPs represent an attractive target of rational therapies to combat vascular inflammatory syndromes. Herein we briefly describe the cellular and molecular etiology of atherosclerosis, and summarize our current understanding of RBPs and their specific roles in regulation of inflammatory mRNA stability. We also detail RBPs as targets of current anti-inflammatory modalities and how this may translate into better treatment for vascular inflammatory diseases.
Collapse
|
247
|
Bilancio A, Rinaldi B, Oliviero MA, Donniacuo M, Monti MG, Boscaino A, Marino I, Friedman L, Rossi F, Vanhaesebroeck B, Migliaccio A. Inhibition of p110δ PI3K prevents inflammatory response and restenosis after artery injury. Biosci Rep 2017; 37:BSR20171112. [PMID: 28851839 PMCID: PMC5617917 DOI: 10.1042/bsr20171112] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 12/13/2022] Open
Abstract
Inflammatory cells play key roles in restenosis upon vascular surgical procedures such as bypass grafts, angioplasty and stent deployment but the molecular mechanisms by which these cells affect restenosis remain unclear. The p110δ isoform of phosphoinositide 3-kinase (PI3K) is mainly expressed in white blood cells. Here, we have investigated whether p110δ PI3K is involved in the pathogenesis of restenosis in a mouse model of carotid injury, which mimics the damage following arterial grafts. We used mice in which p110δ kinase activity has been disabled by a knockin (KI) point mutation in its ATP-binding site (p110δD910A/D910A PI3K mice). Wild-type (WT) and p110δD910A/D910A mice were subjected to longitudinal carotid injury. At 14 and 30 days after carotid injury, mice with inactive p110δ showed strongly decreased infiltration of inflammatory cells (including T lymphocytes and macrophages) and vascular smooth muscle cells (VSMCs), compared with WT mice. Likewise, PI-3065, a p110δ-selective PI3K inhibitor, almost completely prevented restenosis after artery injury. Our data showed that p110δ PI3K plays a main role in promoting neointimal thickening and inflammatory processes during vascular stenosis, with its inhibition providing significant reduction in restenosis following carotid injury. p110δ-selective inhibitors, recently approved for the treatment of human B-cell malignancies, therefore, present a new therapeutic opportunity to prevent the restenosis upon artery injury.
Collapse
Affiliation(s)
- Antonio Bilancio
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Barbara Rinaldi
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "L. Vanvitelli", Naples, Italy
| | - Maria Antonietta Oliviero
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Maria Donniacuo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "L. Vanvitelli", Naples, Italy
| | - Maria Gaia Monti
- Department of Medical Translational Science, University of Naples "Federico II", Naples, Italy
| | - Amedeo Boscaino
- Department of Histopathology, AORN "Cardarelli", Naples, Italy
| | - Irene Marino
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Lori Friedman
- Translational Oncology, Genentech Inc, South San Francisco, CA, U.S.A
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "L. Vanvitelli", Naples, Italy
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Regional Centre for Pharmacovigilance and Pharmaco-epidemiology - University of Campania "L. Vanvitelli", Naples, Italy
| | - Bart Vanhaesebroeck
- Cell Signalling, UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London WC1E 6BT, U.K
| | - Antimo Migliaccio
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
248
|
Fitzpatrick E, Han X, Liu W, Corcoran E, Burtenshaw D, Morrow D, Helt JC, Cahill PA, Redmond EM. Alcohol Reduces Arterial Remodeling by Inhibiting Sonic Hedgehog-Stimulated Stem Cell Antigen-1 Positive Progenitor Stem Cell Expansion. Alcohol Clin Exp Res 2017; 41:2051-2065. [PMID: 28921619 DOI: 10.1111/acer.13499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/11/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cell and molecular mechanisms mediating the cardiovascular effects of alcohol are not fully understood. Our aim was to determine the effect of moderate ethanol (EtOH) on sonic hedgehog (SHh) signaling in regulating possible stem cell antigen-1 positive (Sca1+ ) progenitor stem cell involvement during pathologic arterial remodeling. METHODS Partial ligation or sham operation of the left carotid artery was performed in transgenic Sca1-enhanced green fluorescent protein (eGFP) mice gavaged with or without "daily moderate" EtOH. RESULTS The EtOH group had reduced adventitial thickening and less neointimal formation, compared to ligated controls. There was expansion of eGFP-expressing (i.e., Sca1+ ) cells in remodeled vessels postligation (day 14), especially in the neo intima. EtOH treatment reduced the number of Sca1+ cells in ligated vessel cross-sections concomitant with diminished remodeling, compared to control ligated vessels. Moreover, EtOH attenuated SHh signaling in injured carotids as determined by immunohistochemical analysis of the target genes patched 1 and Gli2, and RT-PCR of whole-vessel Gli2 mRNA levels. Intraperitoneal injection of ligated Sca1-eGFP mice with the SHh signaling inhibitor cyclopamine diminished SHh target gene expression, reduced the number of Sca1+ cells, and ameliorated carotid remodeling. EtOH treatment of purified Sca1+ adventitial progenitor stem cells in vitro inhibited SHh signaling, and their rSHh-induced differentiation to vascular smooth muscle cells. CONCLUSIONS EtOH reduces SHh-responsive Sca1+ progenitor cell myogenic differentiation/expansion in vitro and during arterial remodeling in response to ligation injury in vivo. Regulation of vascular Sca1+ progenitor cells in this way may be an important novel mechanism contributing to alcohol's cardiovascular protective effects.
Collapse
Affiliation(s)
- Emma Fitzpatrick
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Xu Han
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Weimin Liu
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Eoin Corcoran
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Denise Burtenshaw
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - David Morrow
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Jay-Christian Helt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Paul A Cahill
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Eileen M Redmond
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
249
|
Hartman CD, Isenberg BC, Chua SG, Wong JY. Extracellular matrix type modulates cell migration on mechanical gradients. Exp Cell Res 2017; 359:361-366. [PMID: 28821395 PMCID: PMC5603420 DOI: 10.1016/j.yexcr.2017.08.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/21/2017] [Accepted: 08/12/2017] [Indexed: 11/25/2022]
Abstract
Extracellular matrix composition and stiffness are known to be critical determinants of cell behavior, modulating processes including differentiation, traction generation, and migration. Recent studies have demonstrated that the ECM composition can modulate how cells migrate in response to gradients in environmental stiffness, altering a cell's ability to undergo durotaxis. These observations were limited to single varieties of extracellular matrix, but typically cells are exposed to environments containing complex mixtures of extracellular matrix proteins. Here, we investigate migration of NIH 3T3 fibroblasts on mechanical gradients coated with one or more type of extracellular matrix protein. Our results show that NIH 3T3 fibroblasts exhibit durotaxis on fibronectin-coated mechanical gradients but not on those coated with laminin, demonstrating that extracellular matrix type can act as a regulator of cell response to mechanical gradients. Interestingly, NIH 3T3 fibroblasts were also observed to migrate randomly on gradients coated with a mixture of both fibronectin and laminin, suggesting that there may be a complex interplay in the cellular response to mechanical gradients in the presence of multiple extracellular matrix signals. These findings indicate that specific composition of available adhesion ligands is a critical determinant of a cell's migratory response to mechanical gradients.
Collapse
Affiliation(s)
- Christopher D Hartman
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Brett C Isenberg
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Samantha G Chua
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Joyce Y Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States.
| |
Collapse
|
250
|
Jang SA, Park DW, Kwon JE, Song HS, Park B, Jeon H, Sohn EH, Koo HJ, Kang SC. Quinic acid inhibits vascular inflammation in TNF-α-stimulated vascular smooth muscle cells. Biomed Pharmacother 2017; 96:563-571. [PMID: 29032340 DOI: 10.1016/j.biopha.2017.10.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/11/2017] [Accepted: 10/02/2017] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease, and the increased expression of adhesion molecules on vascular smooth muscle cells contributes to the progression of vascular disease. Quinic acid (QA) has been shown to possess radioprotection, anti-neuroinflammatory, and anti-oxidant activities; however, an anti-vascular inflammatory effect has not been reported. This study investigated the effect of QA on the expression of vascular cell adhesion molecule-1 (VCAM-1) stimulated by TNF-α in MOVAS cells. Pre-incubation of MOVAS cells, the mouse vascular smooth muscle cell line for 2h with QA (0.1, 1 and 10 μg/mL) dose-dependently inhibits TNF-α-induced mRNA and protein expression of VCAM-1 and monocyte adhesion. QA inhibits TNF-α-stimulated phosphorylation of MAP kinase and NK-κB activation. Our results indicate that QA inhibits the TNF-α-stimulated induction of VCAM-1 in VSMC by inhibiting the MAP kinase and NF-κB signaling pathways and the adhesion capacity of VSMC, which may explain the ability of QA to inhibit vascular inflammation such as atherosclerosis.
Collapse
Affiliation(s)
- Seon-A Jang
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Dae Won Park
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Jeong Eun Kwon
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hae Seong Song
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Bongkyun Park
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hyelin Jeon
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Eun-Hwa Sohn
- Department of Herbal Medicine Resources, Kangwon National University, Samcheok 25913, Republic of Korea
| | - Hyun Jung Koo
- Department of Medicinal and Industrial Crops, Korea National College of Agriculture and Fisheries, Jeonju 54874, Republic of Korea
| | - Se Chan Kang
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea.
| |
Collapse
|