1
|
Wu H, Li D, Zhang CY, Huang LL, Zeng YJ, Chen TG, Yu K, Meng JW, Lin YX, Guo R, Zhou Y, Gao G. Restoration of ARA metabolic disorders in vascular smooth muscle cells alleviates intimal hyperplasia. Eur J Pharmacol 2024; 983:176824. [PMID: 39265882 DOI: 10.1016/j.ejphar.2024.176824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/14/2024]
Abstract
Intimal hyperplasia (IH) is an innegligible issue for patients undergoing interventional therapy. The proliferation and migration of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor-BB (PDGF-BB) are critical events in the development of IH. While the exact mechanism and effective target for IH needs further investigation. Metabolic disorders of arachidonic acid (ARA) are involved in the occurrence and progression of various diseases. In this study, we found that the expressions of soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) were significantly increased in the VSMCs during balloon injury-induced IH. Then, we employed a COX-2/sEH dual inhibitor PTUPB to increase the concentration of epoxyeicosatrienoic acids (EETs) while prevent the release of pro-inflammatory prostaglandins. Results showed that PTUPB treatment significantly reduced neointimal thickening induced by balloon injury in rats in vivo and inhibited PDGF-BB-induced proliferation and migration of VSMCs in vitro. Our results showed that PTUPB may reverse the phenotypic transition of VSMCs by inhibiting Pttg1 expression. In conclusion, we found that the dysfunction of ARA metabolism in VSMCs contributes to IH, and the COX-2/sEH dual inhibitor PTUPB attenuates IH progression by reversing the phenotypic switch in VSMC through the Sirt1/Pttg1 pathway.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Hyperplasia
- Male
- Rats
- Cyclooxygenase 2/metabolism
- Cell Proliferation/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rats, Sprague-Dawley
- Cell Movement/drug effects
- Epoxide Hydrolases/antagonists & inhibitors
- Epoxide Hydrolases/metabolism
- Tunica Intima/pathology
- Tunica Intima/metabolism
- Tunica Intima/drug effects
- Becaplermin/pharmacology
- Neointima/pathology
- Neointima/metabolism
- Neointima/drug therapy
- Metabolic Diseases/metabolism
- Metabolic Diseases/drug therapy
- Metabolic Diseases/pathology
Collapse
Affiliation(s)
- Hui Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Dai Li
- Phase I Clinical Research Center, Xiangya Hospital, Central South University, Changsha, 410005, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Ling-Li Huang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - You-Jie Zeng
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Tian-Ge Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Ke Yu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jia-Wei Meng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yu-Xin Lin
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, China.
| | - Ge Gao
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
2
|
Zhang N, Song X, Bian Y, Bai R, Yang H, Wang G, Li H, Xiao C. Renin and (pro)renin receptors induce vascular smooth muscle cell proliferation and neointimal hyperplasia by activating oxidative stress and inflammation. Vasc Med 2024; 29:470-482. [PMID: 39212227 DOI: 10.1177/1358863x241261368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Introduction: Renin and prorenin promote the proliferation of vascular smooth muscle cells (VSMCs) through the (pro)renin receptor, or (P)RR, to promote restenosis occurrence. This study aimed to explore whether prorenin promoted the proliferation of VSMCs in a (P)RR-mediated Ang II-independent manner. Methods: Losartan and PD123319 were used to block the interaction between (P)RR and angiotensin in vitro. Cells were treated with renin, platelet-derived growth factor (PDGF), or RNAi-(P)RR, either jointly or individually. Cell proliferation was measured via Cell Counting Kit-8 (CCK-8) and flow cytometry methods; moreover, real-time polymerase chain reaction (RT-PCR) and Western blot (WB) assays were used to detect the expression of cyclin D1, proliferating cell nuclear antigen (PCNA), (P)RR, NOX1, and phosphatidylinositol 3-kinase (PI3K)/AKT signaling proteins. Immunofluorescence staining was conducted to measure the expression of (P)RR, and the levels of renin, PDGF-BB, inflammatory factors, and oxidative stress were determined by using enzyme-linked immunosorbent assay (ELISA). Moreover, a balloon catheter was used to enlarge the carotid artery of the Sprague Dawley rats. PRO20 was applied to identify angiotensin II (Ang II). The hematoxylin and eosin, RT-PCR, and WB results validated the cell assay results. Results: Renin promoted the proliferation of rat VSMCs by enhancing cell viability and cell cycle protein expression when Ang II was blocked, but silencing (P)RR inhibited this effect. Furthermore, renin enhanced NOX1-mediated oxidative stress and inflammation by activating the extracellular signal-regulated kinase 1/2 (ERK1/2)-AKT pathway in vitro. Similarly, the inhibition of (P)RR resulted in the opposite phenomenon. Importantly, the inhibition of (P)RR inhibited neointimal hyperplasia in vivo after common carotid artery injury by restraining NOX1-mediated oxidative stress through the downregulation of the ERK1/2-AKT pathway. The animal study confirmed these findings. Conclusion: Renin and (P)RR induced VSMC proliferation and neointimal hyperplasia by activating oxidative stress, inflammation, and the ERK1/2-AKT pathway in an Ang II-independent manner.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/genetics
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Hyperplasia
- Inflammation Mediators/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Neointima
- Oxidative Stress/drug effects
- Prorenin Receptor
- Proto-Oncogene Proteins c-akt/metabolism
- Rats, Sprague-Dawley
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Renin/metabolism
- Renin-Angiotensin System/drug effects
- Signal Transduction
Collapse
Affiliation(s)
- Nana Zhang
- Department of Hypertension, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaosu Song
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yunfei Bian
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Bai
- Central Lab, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huiyu Yang
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Gang Wang
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hong Li
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chuanshi Xiao
- Department of Cardiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
3
|
Wang M, Li S, Liu H, Liu M, Zhang J, Wu Y, Xiao C, Huang H. Large-conductance Ca 2 +-activated K + channel β1-subunit maintains the contractile phenotype of vascular smooth muscle cells. Front Cardiovasc Med 2022; 9:1062695. [PMID: 36568562 PMCID: PMC9780463 DOI: 10.3389/fcvm.2022.1062695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background Vascular smooth muscle cells (VSMCs) phenotype switching is very important during the pathogenesis and progression of vascular diseases. However, it is not well understood how normal VSMCs maintain the differentiated state. The large-conductance Ca2+-activated K+ (BKCa) channels are widely expressed in VSMCs and regulate vascular tone. Nevertheless, there is limited understanding of the role of the BKCa channel in modulation of the VSMC phenotype. Methods and results We assessed BKCa channel expression levels in normal and injured carotid arteries from rats of the balloon-injury model. A strong decrease of BKCa-β1 was seen in the injured carotid arteries, accompanied by a parallel decrease of the VSMC contractile markers. BKCa-β1 in primary rat aortic VSMCs was decreased with the increase of passage numbers and the stimulation of platelet-derived growth factor (PDGF)-BB. Conversely, transforming growth factor β upregulated BKCa-β1. Meanwhile, the BKCa-β1 level was positively associated with the levels of VSMC contractile proteins. Intravenous injection of PDGF-BB induced downregulation of BKCa-β1 expression in the carotid arteries. Knockdown of BKCa-β1 favored VSMC dedifferentiation, characterized by altered morphology, abnormal actin fiber organization, decreased contractile proteins expression and reduced contractile ability. Furthermore, the resultant VSMC dedifferentiated phenotype rendered increased proliferation, migration, enhanced inflammatory factors levels, and matrix metalloproteinases activity. Studies using primary cultured aortic VSMCs from human recapitulated key findings. Finally, protein level of BKCa-β1 was reduced in human atherosclerotic arteries. Conclusion BKCa-β1 is important in the maintenance of the contractile phenotype of VSMCs. As a novel endogenous defender that prevents pathological VSMC phenotype switching, BKCa-β1 may serve as a potential therapeutic target for treating vascular diseases including post-injury restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Meili Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuanglei Li
- Division of Adult Cardiac Surgery, Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hongshan Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Mingyuan Liu
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jin Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yang Wu
- Division of Adult Cardiac Surgery, Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Cangsong Xiao
- Division of Adult Cardiac Surgery, Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China,Cangsong Xiao,
| | - Haixia Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,*Correspondence: Haixia Huang,
| |
Collapse
|
4
|
Wang X, Gao B, Feng Y. Recent advances in inhibiting atherosclerosis and restenosis: from pathogenic factors, therapeutic agents to nano-delivery strategies. J Mater Chem B 2022; 10:1685-1708. [DOI: 10.1039/d2tb00003b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Due to dominant atherosclerosis etiology, cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide. In clinical trials, advanced atherosclerotic plaques can be removed by angioplasty and vascular...
Collapse
|
5
|
Zhou W, Ye SD, Wang W. Elevated retinol binding protein 4 levels are associated with atherosclerosis in diabetic rats via JAK2/STAT3 signaling pathway. World J Diabetes 2021; 12:466-479. [PMID: 33889291 PMCID: PMC8040077 DOI: 10.4239/wjd.v12.i4.466] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/03/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Atherosclerosis is a major cause of mortality worldwide and is driven by multiple risk factors, including diabetes, which results in an increased atherosclerotic burden, but the precise mechanisms for the occurrence and development of diabetic atherosclerosis have not been fully elucidated.
AIM To summarize the potential role of retinol binding protein 4 (RBP4) in the pathogenesis of diabetic atherosclerosis, particularly in relation to the RBP4-Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway.
METHODS Male Wistar rats were randomly divided into three groups, including a control group (NC group), diabetic rat group (DM group), and diabetic atherosclerotic rat group (DA group). The contents of total cholesterol (TC), high-density lipoprotein cholesterol (HDL-c), triglycerides (TG), low-density lipoprotein cholesterol (LDL-c), fasting insulin (FINS), fasting plasma glucose, and hemoglobin A1c (HbA1c) were measured. Moreover, the adipose and serum levels of RBP4, along with the expression levels of JAK2, phosphorylated JAK2 (p-JAK2), STAT3, phosphorylated STAT3 (p-STAT3), B-cell lymphoma-2 (Bcl-2), and Cyclin D1 in aortic tissues were also measured. Besides, homeostasis model assessment of insulin resistance (HOMA-IR) and atherogenic indexes (AI) were calculated.
RESULTS Compared with the NC and DM groups, the levels LDL-c, TG, TC, FINS, HOMA-IR, RBP4, and AI were upregulated, whereas that of HDL-c was downregulated in the DA group (P < 0.05); the mRNA levels of JAK2, STAT3, Cyclin D1, and Bcl-2 in the DA group were significantly increased compared with the NC group and the DM group; P-JAK2, p-JAK2/JAK2 ratio, p-STAT3, p-STAT3/STAT3 ratio, Cyclin D1, and Bcl-2 at protein levels were significantly upregulated in the DA group compared with the NC group and DM group. In addition, as shown by Pearson analysis, serum RBP4 had a positive correlation with TG, TC, LDL-c, FINS, HbA1C, p-JAK2, p-STAT3, Bcl-2, Cyclin D1, AI, and HOMA-IR but a negative correlation with HDL-c. In addition, multivariable logistic regression analysis showed that serum RBP4, p-JAK2, p-STAT3, and LDL-c were predictors of the presence of diabetic atherosclerosis.
CONCLUSION RBP4 could be involved in the initiation or progression of diabetic atherosclerosis by regulating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Wan Zhou
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
- Laboratory for Diabetes, Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
- Institute of Endocrinology and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Shan-Dong Ye
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
- Laboratory for Diabetes, Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
- Institute of Endocrinology and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Wei Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
- Laboratory for Diabetes, Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
- Institute of Endocrinology and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| |
Collapse
|
6
|
Zhang J, Yang J, Xu C, Hu Q, Hu J, Chen J, Jiang H. Down-regulation of Suv39h1 attenuates neointima formation after carotid artery injury in diabetic rats. J Cell Mol Med 2020; 24:973-983. [PMID: 31736204 PMCID: PMC6933362 DOI: 10.1111/jcmm.14809] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/20/2019] [Accepted: 09/12/2019] [Indexed: 12/22/2022] Open
Abstract
Patients with diabetes have an increased risk of vascular complications. Suv39h1, a histone methyltransferase, plays a protective role against myocardial injury in diabetes. Herein, we intend to explore whether Suv39h1 could affect neointimal formation after vascular injury in diabetic rats and reveal the underlying mechanism. In this study, we generated adenovirus expressing Suv39h1 as well as lentivirus expressing Suv39h1-targeting shRNA and evaluated the significance of Suv39h1 in vascular smooth muscle cells (VSMCs) under diabetic conditions. In vitro, we examined proliferative and migratory behaviours as well as the underlying signalling mechanisms in VSMCs in response to high glucose treatment. In vivo, we induced diabetes in SD rats with streptozocin and established the common carotid artery balloon injury model. Suv39h1 was found to be both necessary and sufficient to promote VSMC proliferation and migration under high glucose conditions. We observed corresponding changes in intracellular signalling molecules including complement C3 and phosphor-ERK1/2. However, either up-regulating or down-regulating Suv39h1, phosphor-p38 level was not significantly affected. Consistently, Suv39h1 overexpression led to accelerated neointima formation, while knocking down Suv39h1 reduced it following carotid artery injury in diabetic rats. Using microarray analyses, we showed that altering the Suv39h1 level in vivo dramatically altered the expression of myriad genes mediating different biological processes and molecular function. This study reveals the novel role of Suv39h1 in VSMCs of diabetes and suggests its potential role as a therapeutic target in diabetic vascular injury.
Collapse
Affiliation(s)
- Jing Zhang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Department of CardiologyThe First College of Clinical Medical ScienceChina Three Gorges University & Yichang Central People's HospitalYichangChina
| | - Jian Yang
- Department of CardiologyThe First College of Clinical Medical ScienceChina Three Gorges University & Yichang Central People's HospitalYichangChina
| | - Changwu Xu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Qi Hu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jun Hu
- Central LaboratoryThe First College of Clinical Medical ScienceChina Three Gorges University & Yichang Central People's HospitalYichangChina
| | - Jing Chen
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hong Jiang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
7
|
Zhang B, Mo X, Yu F, Ma Y, Yan F. Ultrasound monitoring of magnet-guided delivery of mesenchymal stem cells labeled with magnetic lipid–polymer hybrid nanobubbles. Biomater Sci 2020; 8:3628-3639. [PMID: 32529995 DOI: 10.1039/d0bm00473a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells labeled with positively charged magnetic lipid–polymer hybrid nanobubbles could be tracked for magnet-guided delivery onto the site of an injured artery using ultrasound.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Ultrasound in Medicine
- Shanghai East Hospital
- Tongji University School of Medicine
- Shanghai
- China
| | - Xinhai Mo
- Department of Ultrasound in Medicine
- Shanghai East Hospital
- Tongji University School of Medicine
- Shanghai
- China
| | - Fei Yu
- Department of Ultrasound in Medicine
- Shanghai East Hospital
- Tongji University School of Medicine
- Shanghai
- China
| | - Yuqin Ma
- Department of Ultrasound
- Shenzhen Second People's Hospital
- The First Affiliated Hospital of Shenzhen University
- Shenzhen
- China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology
- Shenzhen Institute of Synthetic Biology
- Shenzhen Institutes of Advanced Technology
- Chinese Academy of Sciences
- Shenzhen
| |
Collapse
|
8
|
Luo X, Yang D, Wu W, Long F, Xiao C, Qin M, Law BY, Suguro R, Xu X, Qu L, Liu X, Zhu YZ. Critical role of histone demethylase Jumonji domain-containing protein 3 in the regulation of neointima formation following vascular injury. Cardiovasc Res 2019; 114:1894-1906. [PMID: 29982434 DOI: 10.1093/cvr/cvy176] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/30/2017] [Indexed: 12/31/2022] Open
Abstract
Aims Jumonji domain-containing protein 3 (JMJD3), also called lysine specific demethylase 6B (KDM6b), is an inducible histone demethylase which plays an important role in many biological processes, however, its function in vascular remodelling remains unknown. We aim to demonstrate that JMJD3 mediates vascular neointimal hyperplasia following carotid injury, and proliferation and migration in platelet-derived growth factor BB (PDGF-BB)-induced vascular smooth muscle cells (VSMCs). Methods and results By using both genetic and pharmacological approaches, our study provides the first evidence that JMJD3 controls PDGF-BB-induced VSMCs proliferation and migration. Furthermore, our in vivo mouse and rat intimal thickening models demonstrate that JMJD3 is a novel mediator of neointima formation based on its mediatory effects on VSMCs proliferation, migration, and phenotypic switching. We further show that JMJD3 ablation by small interfering RNA or inhibitor GSK J4 can suppress the expression of NADPH oxidase 4 (Nox4), which is correlated with H3K27me3 enrichment around the gene promoters. Besides, deficiency of JMJD3 and Nox4 prohibits autophagic activation, and subsequently attenuates neointima and vascular remodelling following carotid injury. Above all, the increased expression of JMJD3 and Nox4 is further confirmed in human atherosclerotic arteries plaque specimens. Conclusions JMJD3 is a novel factor involved in vascular remodelling. Deficiency of JMJD3 reduces neointima formation after vascular injury by a mechanism that inhibits Nox4-autophagy signalling activation, and suggesting JMJD3 may serve as a perspective target for the prevention and treatment of vascular diseases.
Collapse
Affiliation(s)
- XiaoLing Luo
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - Di Yang
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - WeiJun Wu
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - Fen Long
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - ChenXi Xiao
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - Ming Qin
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - Betty YuenKwan Law
- State Key Laboratory of Quality Research, Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Rinkiko Suguro
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - Xin Xu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; and
| | - LeFeng Qu
- Department of Vascular Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - XinHua Liu
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China
| | - Yi Zhun Zhu
- Department of Pharmacology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Pharmacy, Fudan University, 826, Zhangheng Road, Shanghai, China.,State Key Laboratory of Quality Research, Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
9
|
Di Stolfo G, Mastroianno S, Ruggieri M, Fontana A, Marinucci R, Copetti M, Minervini MM, Savino L, Mastroianno M, Savino M, Pacilli MA, Di Mauro L, Potenza DR, Cascavilla N, Paroni G, Russo A. Timing of clopidogrel loading dose on peripheral blood endothelial progenitor cells, SDF-1α and neointimal hyperplasia in carotid stenting. Clin Hemorheol Microcirc 2019; 72:23-38. [DOI: 10.3233/ch-180429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Giuseppe Di Stolfo
- Cardiology Unit, Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Sandra Mastroianno
- Cardiology Unit, Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Maurizio Ruggieri
- Vascular Surgery Unit, Cardiolovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Andrea Fontana
- Unit of Biostatistics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Roberto Marinucci
- Vascular Surgery Unit, Cardiolovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Massimiliano Copetti
- Unit of Biostatistics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Maria Marta Minervini
- Hematology Unit, Onco-hematology Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Lucia Savino
- Hematology Unit, Onco-hematology Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Mario Mastroianno
- Unit of Information Systems and Innovation Research, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Maria Savino
- Transfusion Medicine Unit and Laboratory of Clinical Chemistry, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Michele Antonio Pacilli
- Cardiology Unit, Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Lazzaro Di Mauro
- Transfusion Medicine Unit and Laboratory of Clinical Chemistry, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Domenico Rosario Potenza
- Cardiology Unit, Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Nicola Cascavilla
- Hematology Unit, Onco-hematology Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Giovanni Paroni
- Vascular Surgery Unit, Cardiolovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Aldo Russo
- Cardiology Unit, Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| |
Collapse
|
10
|
Bio-Based Covered Stents: The Potential of Biologically Derived Membranes. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:135-151. [DOI: 10.1089/ten.teb.2018.0207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11
|
Du JB, Zhang W, Li N, Jiang H, Liu Y, Gao J, Chen ST, Cong HL, Wei YL. Association study of matrix metalloproteinase 3 5A/6A polymorphism with in-stent restenosis after percutaneous coronary interventions in a Han Chinese population. J Int Med Res 2019; 48:300060519827145. [PMID: 30732526 PMCID: PMC7140217 DOI: 10.1177/0300060519827145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objective We aimed to investigate the association between the 5A/6A promoter
polymorphism in the matrix metalloproteinase 3 (MMP3) gene
and in-stent restenosis (ISR) in a regional Chinese population. Methods A total of 818 patients who underwent primary implantation of drug-eluting
stents were enrolled and received a 6-month follow-up angiography and DNA
genotyping of the 5A/6A polymorphism. Results ISR was found in 36.9% of all patients (302 ISR vs. 516 no ISR). The genotype
proportion of 6A6A was significantly increased in ISRs (74.2% ISR vs. 66.8%
no ISR), whereas the allele frequency of 5A was significantly decreased in
ISR patients (25.8%) compared with controls who did not undergo ISR
(33.1%). Conclusions Our data indicate that the MMP3 6A6A genotype is a genetic
susceptibility factor for ISR after coronary stent placement, but the 5A
allele can lower the risk for patients within 6 months after stenting.
Therefore, genotyping 5A/6A in the MMP3 promoter is
suggested for patients who undergo coronary stent implantation.
Collapse
Affiliation(s)
- Ji-Bing Du
- Department of Cardiovascular Medicine, Tianjin Cardiovascular Disease Institute, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Wei Zhang
- Department of Immunology, Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, People's Republic of China.,Department of Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Na Li
- Department of Pathology, Xingtai Medical College, Xingtai, Hebei, People's Republic of China
| | - Hua Jiang
- Department of Cardiovascular Medicine, Tianjin Cardiovascular Disease Institute, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Yin Liu
- Department of Cardiovascular Medicine, Tianjin Cardiovascular Disease Institute, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Jing Gao
- Department of Cardiovascular Medicine, Tianjin Cardiovascular Disease Institute, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Shu-Tao Chen
- Department of Cardiovascular Medicine, Tianjin Cardiovascular Disease Institute, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Hong-Liang Cong
- Department of Cardiovascular Medicine, Tianjin Cardiovascular Disease Institute, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Yi-Liang Wei
- Department of Immunology, Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
12
|
Pearce C, Islam N, Bryce R, McNair ED. Advanced Glycation End Products:Receptors for Advanced Glycation End Products Axis in Coronary Stent Restenosis: A Prospective Study. Int J Angiol 2018; 27:213-222. [PMID: 30410293 DOI: 10.1055/s-0038-1673660] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
This study was aimed to correlate the pre- and 6-month postpercutaneous coronary intervention (PCI) serum concentrations of advanced glycation end products (AGE), soluble receptors for advanced glycation end products (sRAGE), AGE/sRAGE ratio, and serum malondialdehyde (MDA) levels with in-stent restenosis (ISR) among patients receiving either a drug-eluting stent (DES) or a bare-metal stent (BMS).In-stent restenosis remains as an adverse outcome following PCI. Sixty consecutive nondiabetic, Caucasian male patients, diagnosed with a non-ST-elevation myocardial infarction who received either a DES or BMS via PCI, were enrolled. Baseline levels of serum AGE, sRAGE, AGE/sRAGE ratios, MDA, and angiographic parameters were determined at stenting and at 6 months. Patients with and without ISR at 6 months were compared on both baseline and 6-month biomarker levels and within stent types.The pre-PCI serum AGE levels and AGE/sRAGE ratios were higher in ISR patients compared with non-ISR patients, while the pre-PCI and post-PCI serum sRAGE levels were lower in ISR patients compared with non-ISR patients. The pre and post-PCI levels of MDA were also higher in ISR patients. Comparing stent types, relative levels of MDA between those with and without ISR at the respective time points were similar, although changes between time points appeared type specific.Post-PCI ISR correlates with low serum values of sRAGE and high serum values of AGE, MDA, and AGE/sRAGE ratio which are present at stenting. The associations of baseline AGE, sRAGE, AGE/sRAGE, and MDA levels with ISR appear consistent between stent types.
Collapse
Affiliation(s)
- Colin Pearce
- Division of Cardiology, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Naorin Islam
- Department of Community Health and Epidemiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Robyn Bryce
- Department of Community Health and Epidemiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Erick Donnell McNair
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
13
|
Song T, Zhao J, Jiang T, Jin X, Li Y, Liu X. Formononetin protects against balloon injury‑induced neointima formation in rats by regulating proliferation and migration of vascular smooth muscle cells via the TGF‑β1/Smad3 signaling pathway. Int J Mol Med 2018; 42:2155-2162. [PMID: 30066831 DOI: 10.3892/ijmm.2018.3784] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/25/2018] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the effects of formononetin (FMN) against balloon injury‑induced neointima formation in vivo and platelet‑derived growth factor (PDGF)‑BB‑induced proliferation and migration of vascular smooth muscle cells (VSMCs) in vitro, and explored the underlying mechanisms. A rat model of carotid artery injury was established, in order to examine the effects of FMN on balloon injury‑induced neointima formation. Histological observation of the carotid artery tissues was conducted by hematoxylin and eosin staining. VSMC proliferation during neointima formation was observed by proliferating cell nuclear antigen staining. Subsequently, rat aortic VSMCs were isolated, and the effects of FMN on PDGF‑BB‑induced VSMC proliferation and migration were determined using Cell Counting Kit‑8 and Transwell/wound healing assays, respectively. Immunohistochemical and immunocytochemical staining was applied to measure the expression of transforming growth factor (TGF)‑β in carotid artery tissues and VSMCs, respectively. SMAD family member 3 (Smad3)/phosphorylated (p)‑Smad3 expression was examined by western blotting. FMN treatment significantly inhibited the abnormal proliferation of smooth muscle cells in neointima, and alterations to the vascular structure were attenuated. In addition, pretreatment with FMN effectively inhibited the proliferation of PDGF‑BB‑stimulated VSMCs (P<0.05). FMN also reduced the number of cells that migrated to the lower surface of the Transwell chamber and decreased wound‑healing percentage (P<0.05). The expression levels of TGF‑β were decreased by FMN treatment in vivo and in vitro, and Smad3/p‑Smad3 expression was also markedly inhibited. In conclusion, FMN significantly protected against balloon injury‑induced neointima formation in the carotid artery of a rat model; this effect may be associated with the regulation of VSMC proliferation and migration through altered TGF‑β1/Smad3 signaling.
Collapse
Affiliation(s)
- Tao Song
- Department of Vascular Surgery, Linyi Peoples' Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| | - Jingdong Zhao
- Department of Vascular Surgery, Linyi Peoples' Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| | - Tongbai Jiang
- Department of Vascular Surgery, Linyi Peoples' Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| | - Xing Jin
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yubin Li
- Department of Vascular Surgery, Linyi Peoples' Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| | - Xinrong Liu
- Hemodialysis Center, Linyi Peoples' Hospital Affiliated to Shandong University, Linyi, Shandong 276003, P.R. China
| |
Collapse
|
14
|
de Bruin RG, Rabelink TJ, van Zonneveld AJ, van der Veer EP. Emerging roles for RNA-binding proteins as effectors and regulators of cardiovascular disease. Eur Heart J 2018; 38:1380-1388. [PMID: 28064149 DOI: 10.1093/eurheartj/ehw567] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/02/2016] [Indexed: 12/18/2022] Open
Abstract
The cardiovascular system comprises multiple cell types that possess the capacity to modulate their phenotype in response to acute or chronic injury. Transcriptional and post-transcriptional mechanisms play a key role in the regulation of remodelling and regenerative responses to damaged cardiovascular tissues. Simultaneously, insufficient regulation of cellular phenotype is tightly coupled with the persistence and exacerbation of cardiovascular disease. Recently, RNA-binding proteins such as Quaking, HuR, Muscleblind, and SRSF1 have emerged as pivotal regulators of these functional adaptations in the cardiovascular system by guiding a wide-ranging number of post-transcriptional events that dramatically impact RNA fate, including alternative splicing, stability, localization and translation. Moreover, homozygous disruption of RNA-binding protein genes is commonly associated with cardiac- and/or vascular complications. Here, we summarize the current knowledge on the versatile role of RNA-binding proteins in regulating the transcriptome during phenotype switching in cardiovascular health and disease. We also detail existing and potential DNA- and RNA-based therapeutic approaches that could impact the treatment of cardiovascular disease in the future.
Collapse
Affiliation(s)
- Ruben G de Bruin
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Ton J Rabelink
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| | - Eric P van der Veer
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands.,Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2300RC, The Netherlands
| |
Collapse
|
15
|
Ma X, Jiang C, Li Y, Feng L, Liu J, Wang J. Inhibition effect of tacrolimus and platelet-derived growth factor-BB on restenosis after vascular intimal injury. Biomed Pharmacother 2017. [PMID: 28633129 DOI: 10.1016/j.biopha.2017.06.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Excessive proliferation and migration of vascular smooth muscle cells(VSMCs) and delayed proliferation and migration of endothelial cells(ECs) were the main cause of restenosis after endovascular interventional therapy. Since tacrolimus has proved to be more sensitive to inhibiting VSMCs' proliferation,and platelet-derived growth factor-BB(PDGF-BB) benefitted ECs' and VSMCs' proliferation, this study was aimed to identify combined effect of tacrolimus and PDGF-BB, investigate any mechanisms underneath and demonstrate combined effect of two drugs in vivo. As the results showed we confirmed differential effect of PDGF-BB and tacrolimus on ECs and VSMCs. On the concentration level of 2-5μg/ml tacrolimus plus 10ng/ml PDGF-BB, combination of drugs could effectively promote ECs proliferation and migration, and meanwhile inhibit VSMCs proliferation and migration, and the inhibition of p-mTOR's expression within VSMCs played an important role in this differentiated effect. Raising concentration level of PDGF-BB would weaken inhibitory effect of tacrolimus on both kinds of cell. For injured intima, the mix solution of two drugs could promote intima healing and suppress excessive intimal hyperplasia.
Collapse
Affiliation(s)
- Xu Ma
- Department of Interventional Radiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, No 600 Yishan road, Xuhui District, Shanghai, 200233, China
| | - Chunyu Jiang
- Department of Interventional Radiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, No 600 Yishan road, Xuhui District, Shanghai, 200233, China
| | - Yuehua Li
- Department of Interventional Radiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, No 600 Yishan road, Xuhui District, Shanghai, 200233, China
| | - Lishuai Feng
- Department of Interventional Radiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, No 600 Yishan road, Xuhui District, Shanghai, 200233, China
| | - Jingjing Liu
- Department of Interventional Radiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, No 600 Yishan road, Xuhui District, Shanghai, 200233, China
| | - Jianbo Wang
- Department of Interventional Radiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, No 600 Yishan road, Xuhui District, Shanghai, 200233, China.
| |
Collapse
|
16
|
Ichihashi S, Wolf F, Schmitz-Rode T, Kichikawa K, Jockenhoevel S, Mela P. In Vitro Quantification of Luminal Denudation After Crimping and Balloon Dilatation of Endothelialized Covered Stents. Cardiovasc Intervent Radiol 2017; 40:1229-1236. [DOI: 10.1007/s00270-017-1661-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/21/2017] [Indexed: 10/19/2022]
|
17
|
Lee KP, Kim JE, Kim H, Chang HR, Lee DW, Park WH. Bo-Gan-Whan regulates proliferation and migration of vascular smooth muscle cells. Altern Ther Health Med 2016; 16:306. [PMID: 27549769 PMCID: PMC4994174 DOI: 10.1186/s12906-016-1292-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 08/17/2016] [Indexed: 02/05/2023]
Abstract
Background Bo-Gan-Whan (BGH), a Korean polyherbal medicine, is used as a hepatoprotective drug. It has six natural sources, and has been demonstrated to have anti-oxidative, anti-cancer, and anti-inflammatory properties; however, its effect on vascular diseases remains unclear. Methods Cell viability and proliferation assays were employed using an EZ-Cytox Cell Viability Assay Kit. Platelet-derived growth factor (PDGF)-BB-induced vascular smooth muscle cell (VSMC) migration was measured by scratch wound healing assay and Boyden chamber assay. The expression levels of the phosphorylated signaling proteins relevant to proliferation, including extracellular signal-regulated kinase (ERK) 1/2 and p38 mitogen-activated protein kinase (MAPK) were determined by western blot analysis. Chromatogram and mass analysis were employed by Ultra Performance Liquid Chromatography (UPLC) system. Cell prolife ration and migration were also explored using the PDGF-BB-induced aortic sprout assay. Results BGH (100–500 μg/mL) significantly inhibited the proliferation and migration of PDGF-BB-stimulated VSMCs through the reduced phosphorylation of ERK1/2 and p38 MAPK in comparison to untreated PDGF-BB-stimulated VSMC. Moreover, we identified the paeoniflorin as the major composition of BGH. Conclusions We suggest that BGH may have an anti-atherosclerosis effect by inhibiting the proliferation and migration of PDGF-BB-stimulated VSMCs through down-regulation of ERK1/2 and p38 MAPK phosphorylation.
Collapse
|
18
|
Zhou X, Mou Y, Shen X, Yang T, Liu J, Liu F, Dong J, Liao L. The role of atorvastatin on the restenosis process post-PTA in a diabetic rabbit model. BMC Cardiovasc Disord 2016; 16:153. [PMID: 27422557 PMCID: PMC4947282 DOI: 10.1186/s12872-016-0324-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 06/10/2016] [Indexed: 12/01/2022] Open
Abstract
Background Restenosis remains to be a major limitation of percutaneous transluminal angioplasty (PTA) for diabetic patients with peripheral vascular disease (PVD). Despite of stations routine implements to prevent such progress, its exact effect is unclear. Methods and results In our study, balloon was successfully implanted in the iliac artery of atherosclerotic rabbit. Patency of the narrowed artery was interrogated using ultrasound. Atorvastatin or vehicle was administered orally to rabbits from day 0 to day 28 after double-injury surgery. On day 7, day 14, and day 28, restenotic arteries were harvested and processed for histopathlogical analysis. Our data show that, after double-injury surgery, the intima was composed mostly by SMCs at all time course in rabbits undergoing surgery process. Significant increases in stenosis rates were noted from day 7 to day 14 (from 21 ± 5.85 % to 60.93 ± 12.46 %). On day 28 after double-injury surgery, severe restenosis was observed and daily administration of atorvastatin cannot prevent restenosis’ formation (88.69 ± 3.71 % vs. 90.02 ± 3.11 %, P > 0.05). The PCNA index and SMCs proliferation were correlated with the scores of the vascular pathology. Conclusions Our results indicate that double-injury model can mimic clinical restenosis, based on this model, atorvastatin showed no therapeutic effect on restenosis process in diabetic rabbits after PTA.
Collapse
Affiliation(s)
- Xiaojun Zhou
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766, Jingshi Road, Lixia District, Jinan, 250000, Shandong Province, China
| | - Yaru Mou
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Shandong University, Jinan, Shandong, China
| | - Xue Shen
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianshu Yang
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ju Liu
- Laboratory of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Fupeng Liu
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766, Jingshi Road, Lixia District, Jinan, 250000, Shandong Province, China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital of Shandong University, No.44, wenhuan Road, Lixia District, Jinan, 250000, Shandong Province, China.
| | - Lin Liao
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766, Jingshi Road, Lixia District, Jinan, 250000, Shandong Province, China.
| |
Collapse
|
19
|
Nrf2/Keap1 system regulates vascular smooth muscle cell apoptosis for vascular homeostasis: role in neointimal formation after vascular injury. Sci Rep 2016; 6:26291. [PMID: 27198574 PMCID: PMC4873803 DOI: 10.1038/srep26291] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/28/2016] [Indexed: 01/07/2023] Open
Abstract
Abnormal increases in vascular smooth muscle cells (VSMCs) in the intimal region after a vascular injury is a key event in developing neointimal hyperplasia. To maintain vascular function, proliferation and apoptosis of VSMCs is tightly controlled during vascular remodeling. NF-E2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1) system, a key component of the oxidative stress response that acts in maintaining homeostasis, plays an important role in neointimal hyperplasia after a vascular injury; however, the role of Nrf2/Keap1 in VSMC apoptosis has not been clarified. Here we report that 14 days after arterial injury in mice, TUNEL-positive VSMCs are detected in both the neointimal and medial layers. These layers contain cells expressing high levels of Nrf2 but low Keap1 expression. In VSMCs, Keap1 depletion induces features of apoptosis, such as positive TUNEL staining and annexin V binding. These changes are associated with an increased expression of nuclear Nrf2. Simultaneous Nrf2 depletion inhibits Keap1 depletion-induced apoptosis. At 14 days after the vascular injury, Nrf2-deficient mice demonstrated fewer TUNEL-positive cells and increased neointimal formation in the neointimal and medial areas. The results suggest that the Nrf2/Keap1 system regulates VSMC apoptosis during neointimal formation, thereby inhibiting neointimal hyperplasia after a vascular injury.
Collapse
|
20
|
Vannozzi L, Ricotti L, Filippeschi C, Sartini S, Coviello V, Piazza V, Pingue P, La Motta C, Dario P, Menciassi A. Nanostructured ultra-thin patches for ultrasound-modulated delivery of anti-restenotic drug. Int J Nanomedicine 2015; 11:69-91. [PMID: 26730191 PMCID: PMC4694686 DOI: 10.2147/ijn.s92031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
This work aims to demonstrate the possibility to fabricate ultra-thin polymeric films loaded with an anti-restenotic drug and capable of tunable drug release kinetics for the local treatment of restenosis. Vascular nanopatches are composed of a poly(lactic acid) supporting membrane (thickness: ~250 nm) on which 20 polyelectrolyte bilayers (overall thickness: ~70 nm) are alternatively deposited. The anti-restenotic drug is embedded in the middle of the polyelectrolyte structure, and released by diffusion mechanisms. Nanofilm fabrication procedure and detailed morphological characterization are reported here. Barium titanate nanoparticles (showing piezoelectric properties) are included in the polymeric support and their role is investigated in terms of influence on nanofilm morphology, drug release kinetics, and cell response. Results show an efficient drug release from the polyelectrolyte structure in phosphate-buffered saline, and a clear antiproliferative effect on human smooth muscle cells, which are responsible for restenosis. In addition, preliminary evidences of ultrasound-mediated modulation of drug release kinetics are reported, thus evaluating the influence of barium titanate nanoparticles on the release mechanism. Such data were integrated with quantitative piezoelectric and thermal measurements. These results open new avenues for a fine control of local therapies based on smart responsive materials.
Collapse
Affiliation(s)
- Lorenzo Vannozzi
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera, Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera, Italy
| | - Carlo Filippeschi
- Center for MicroBioRobotics at SSSA, Istituto Italiano di Tecnologia, Pontedera, Italy
| | | | - Vito Coviello
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Vincenzo Piazza
- Center for Nanotechnology Innovation at NEST, Istituto Italiano di Tecnologia, Pisa, Italy
| | | | | | - Paolo Dario
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera, Italy
| | - Arianna Menciassi
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera, Italy
| |
Collapse
|
21
|
Zhou X, Dong J, Zhang L, Liu J, Dong X, Yang Q, Liu F, Liao L. Hyperglycemia has no effect on development of restenosis after percutaneous transluminal angioplasty (PTA) in a diabetic rabbit model. J Endocrinol 2015; 224:119-25. [PMID: 25385870 DOI: 10.1530/joe-14-0391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It is well known that hyperglycemia is a trigger of atherosclerosis in patients with diabetes mellitus. However, the role of hyperglycemia in restenosis remains unclear. In this study, we investigated the effects of hyperglycemia on restenosis. Stenosis was evaluated in two sets of diabetic rabbit models: i) diabetic restenosis versus nondiabetic restenosis and ii) diabetic atherosclerosis versus nondiabetic atherosclerosis. Our results indicated that there was no difference in rates of stenosis between the diabetic and the nondiabetic groups in restenosis rabbit models. However, the incidence of stenosis was significantly higher in the diabetic atherosclerosis group compared with the nondiabetic atherosclerosis group. Similarly, the intima-media thickness and cell proliferation rate were significantly increased in the diabetic atherosclerosis group compared with the nondiabetic atherosclerosis group, but there was no difference between the diabetic restenosis and the nondiabetic restenosis groups. Our results indicate that hyperglycemia is an independent risk factor for atherosclerosis, but it has no evident effect on restenosis. These findings indicate that the processes of atherosclerosis and restenosis may involve different pathological mechanisms.
Collapse
Affiliation(s)
- Xiaojun Zhou
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jianjun Dong
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Li Zhang
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Ju Liu
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Xiaofeng Dong
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Qing Yang
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Fupeng Liu
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Lin Liao
- Department of EndocrinologyShandong Provincial Qianfoshan Hospital, Shandong University, No. 16766, Jingshi Road, Lixia District, Jinan, Shandong Province, ChinaDepartment of EndocrinologyQilu Hospital of Shandong University, Jinan, Shandong, ChinaDepartment of SonographyLaboratory of Microvascular MedicineMedical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, ChinaDepartment of Hepatobiliary Surgerythe People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
22
|
Tian L, Ni J, Guo T, Liu J, Dang Y, Guo Q, Zhang L. TSH stimulates the proliferation of vascular smooth muscle cells. Endocrine 2014; 46:651-8. [PMID: 24452868 DOI: 10.1007/s12020-013-0135-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/22/2013] [Indexed: 01/04/2023]
Abstract
Subclinical hypothyroidism (SCH) was reported to be associated with atherosclerosis (AS) in recent studies. Thyroid hormone levels are normal in patients with SCH, but the levels of thyroid-stimulating hormone (TSH) are increased. Thyroid-stimulating hormone receptor (TSHR) in extra-thyroidal tissues plays a pathophysiological role in these conditions. Our previous results demonstrated that TSHR was functional in hepatocytes and revealed elevated total cholesterol levels in the serum, which were an independent risk factor for AS. TSHR is expressed in vascular smooth muscle cells (VSMCs), and VSMC proliferation plays an important role in the development of AS. Cell proliferation was measured by the MTT assay. Intracellular cyclic AMP (cAMP) was measured using a cAMP ELISA kit. Cells were analyzed using a flow cytometer to determine the cell-cycle phase of each cell. For the purpose of detecting cyclin A and cyclin D, immunohistochemical staining and western blots were performed. Real-time PCR was used to assess the VSMC phenotypes. TSH increased cell progression into the G2/M phases and induced VSMC proliferation; thus, functional TSHR was present on VSMCs. Furthermore, the expression of cyclin D1 and cyclin A was increased. In addition, the results indicated that VSMCs undergo a phenotypic transformation from a contractile state to a synthetic state after treatment with different concentrations of TSH. Elevated TSH can promote VSMC proliferation through the cAMP-dependent pathway.
Collapse
Affiliation(s)
- Limin Tian
- Department of Endocrinology, Gansu Provincial Hospital, 204 Dong Gang West Road, Lanzhou, 730000, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Ran F, Liu C, Liu Z, Shang T, Zhou M, Qiao T. Preventive effects of basic fibroblast growth factor on vascular restenosis after balloon angioplasty. Exp Ther Med 2014; 7:1193-1196. [PMID: 24940410 PMCID: PMC3991498 DOI: 10.3892/etm.2014.1562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 01/29/2014] [Indexed: 12/28/2022] Open
Abstract
The aim of the present study was to investigate whether chronic administration of basic fibroblast growth factor (bFGF) following angioplasty in a dog model of atherosclerotic iliac stenosis may restore endothelium function and prevent restenosis (RS). In total, 40 dogs with atherosclerotic stenosis of the right iliac arteries were used in the study. A total of 20 dogs underwent histological examination of the lumen areas prior to (n=10) and immediately following angioplasty (n=10). Intravenous bFGF was administered to 10 dogs (bFGF group) and an additional 10 dogs received vehicle injection (control group). Animals in the two groups were sacrificed 42 days following surgery for in vitro analysis of vascular reactivity and morphometric assessment of the histological cross-sectional areas. The bFGF group exhibited significantly greater maximal endothelium-dependent acetylcholine-induced relaxation (Emax, 43±9%) when compared with the control group (Emax, 8±6%; P<0.05). In addition, the maximal endothelium-independent response of the bFGF group to sodium nitroprusside (Emax, 90±2%) was greater than that of the control group (Emax, 60±2%; P<0.05). Six weeks following angioplasty, the lumen area in the bFGF group (2.01±0.78 mm2) was greater compared with the control group (1.0±0.10%). The lumen area decreased by 58% between immediately after angioplasty and the control group six weeks following angioplasty. Therefore, the results of the present study indicated that administration of bFGF may not only restore endothelium-dependent and -independent relaxation, but also prevent RS in dogs that have undergone angioplasty.
Collapse
Affiliation(s)
- Feng Ran
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Changjian Liu
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Zhao Liu
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Tao Shang
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Min Zhou
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Tong Qiao
- Department of Vascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
24
|
Zhang J, Chen J, Xu C, Yang J, Guo Q, Hu Q, Jiang H. Resveratrol inhibits phenotypic switching of neointimal vascular smooth muscle cells after balloon injury through blockade of Notch pathway. J Cardiovasc Pharmacol 2014; 63:233-239. [PMID: 24603118 DOI: 10.1097/fjc.0000000000000040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Phenotypic switching of vascular smooth muscle cells (VSMCs) plays an initial role in neointimal hyperplasia, the main cause of many occlusive vascular diseases. The aim of this study was to measure the effects of resveratrol (RSV) on the phenotypic transformation of VSMCs and to investigate its mechanism of action. METHODS Cultured VSMCs isolated from rat thoracic aorta were prepared with serum starvation for 72 hours followed by RSV treatment (50-200 μmol/L) and 10% serum stimulation. Male Sprague-Dawley rats, subjected to carotid arteries injury from a balloon catheter, were exposed to intraperitoneal injection of RSV (1 mg/kg) or saline and were killed after 7 or 28 days. RESULTS Compared with cells in the serum-induced group, VSMCs in the RSV or N-[N-(3, 5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) treatment group exhibited significant decreases of proliferation and migration. The total and cytoplasmic Notch-1 levels were declined by RSV, accompanied by a significant increase in smooth muscle α-actin and smooth muscle myosin heavy chain protein. The expression of Notch-1, Jagged-1, Hey-1, and Hey-2 mRNA in balloon-injured arteries at 7 days was decreased by RSV treatment. Arteries from RSV-treated rats showed less neointimal hyperplasia, lower collagen content, and a lower rate of cells positive for proliferating cell nuclear antigen 28 days after injury, compared with saline controls. CONCLUSIONS The results indicate that RSV can attenuate phenotypic switching of VSMCs after arterial injury through inhibition of the Notch pathway.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Carotid Arteries/drug effects
- Carotid Arteries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Cells, Cultured
- Disease Models, Animal
- Hyperplasia/prevention & control
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Neointima/prevention & control
- Rats
- Rats, Sprague-Dawley
- Receptor, Notch1/genetics
- Resveratrol
- Stilbenes/pharmacology
- Time Factors
Collapse
Affiliation(s)
- Jing Zhang
- *Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; †Departments of Cardiology; and ‡Ophthalmology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Lin CM, Hou SW, Wang BW, Ong JR, Chang H, Shyu KG. Molecular mechanism of (-)-epigallocatechin-3-gallate on balloon injury-induced neointimal formation and leptin expression. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:1213-1220. [PMID: 24410132 DOI: 10.1021/jf404479x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Leptin contributes to the pathogenesis of vascular repair and cardiovascular events. This study evaluated the molecular mechanism of EGCG in balloon injury-induced leptin expression. According to immunohistochemical and confocal analyses, leptin expression was increased and the aortic lumen exhibited narrowing after balloon injury. EGCG treatment attenuated leptin expression and diminished neointimal formation. The in vitro study showed that angiotensin II (Ang II) induced the migration and proliferation of cultured vascular smooth muscle cells (VSMCs), whereas treatment with EGCG, leptin siRNA, and c-Jun siRNA inhibited the migration and proliferation of VSMCs significantly. The EMSA shows that balloon injury increased AP-1-binding activity, and EGCG and c-Jun siRNA inhibited the AP-1-binding activity. Western blot and real-time RT-PCR analyses revealed similar results in intimal tissue samples. In summary, balloon injury induces leptin expression in the carotid artery of rats, and EGCG inhibits leptin expression through the JNK/AP-1 pathway and also attenuates neointimal formation.
Collapse
Affiliation(s)
- Chiu-Mei Lin
- Department of Emergency Medicine, Shin Kong Wu Ho-Su Memorial Hospital , Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
26
|
Zhang J, Chen J, Yang J, Xu C, Ding J, Yang J, Guo Q, Hu Q, Jiang H. Sodium ferulate inhibits neointimal hyperplasia in rat balloon injury model. PLoS One 2014; 9:e87561. [PMID: 24489938 PMCID: PMC3906191 DOI: 10.1371/journal.pone.0087561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/23/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND/AIM Neointimal formation after vessel injury is a complex process involving multiple cellular and molecular processes. Inhibition of intimal hyperplasia plays an important role in preventing proliferative vascular diseases, such as restenosis. In this study, we intended to identify whether sodium ferulate could inhibit neointimal formation and further explore potential mechanisms involved. METHODS Cultured vascular smooth muscle cells (VSMCs) isolated from rat thoracic aorta were pre-treated with 200 µmol/L sodium ferulate for 1 hour and then stimulated with 1 µmol/L angiotensin II (Ang II) for 1 hour or 10% serum for 48 hours. Male Sprague-Dawley rats subjected to balloon catheter insertion were administrated with 200 mg/kg sodium ferulate (or saline) for 7 days before sacrificed. RESULTS In presence of sodium ferulate, VSMCs exhibited decreased proliferation and migration, suppressed intracellular reactive oxidative species production and NADPH oxidase activity, increased SOD activation and down-regulated p38 phosphorylation compared to Ang II-stimulated alone. Meanwhile, VSMCs treated with sodium ferulate showed significantly increased protein expression of smooth muscle α-actin and smooth muscle myosin heavy chain protein. The components of Notch pathway, including nuclear Notch-1 protein, Jagged-1, Hey-1 and Hey-2 mRNA, as well as total β-catenin protein and Cyclin D1 mRNA of Wnt signaling, were all significantly decreased by sodium ferulate in cells under serum stimulation. The levels of serum 8-iso-PGF2α and arterial collagen formation in vessel wall were decreased, while the expression of contractile markers was increased in sodium ferulate treated rats. A decline of neointimal area, as well as lower ratio of intimal to medial area was observed in sodium ferulate group. CONCLUSION Sodium ferulate attenuated neointimal hyperplasia through suppressing oxidative stress and phenotypic switching of VSMCs.
Collapse
MESH Headings
- Angioplasty, Balloon/adverse effects
- Angiotensin II/physiology
- Animals
- Carotid Arteries/drug effects
- Carotid Arteries/pathology
- Carotid Artery Diseases/drug therapy
- Carotid Artery Diseases/etiology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Coumaric Acids/pharmacology
- Drug Evaluation, Preclinical
- Hyperplasia/prevention & control
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Neointima/drug therapy
- Neointima/etiology
- Oxidative Stress/drug effects
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
Collapse
Affiliation(s)
- Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, PR China
| | - Changwu Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Jiawang Ding
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, PR China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, PR China
| | - Qing Guo
- Department of Ophthalmology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Qi Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| |
Collapse
|
27
|
Berra-Romani R, Avelino-Cruz JE, Raqeeb A, Della Corte A, Cinelli M, Montagnani S, Guerra G, Moccia F, Tanzi F. Ca²⁺-dependent nitric oxide release in the injured endothelium of excised rat aorta: a promising mechanism applying in vascular prosthetic devices in aging patients. BMC Surg 2013; 13 Suppl 2:S40. [PMID: 24266895 PMCID: PMC3851245 DOI: 10.1186/1471-2482-13-s2-s40] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background Nitric oxide is key to endothelial regeneration, but it is still unknown whether endothelial cell (EC) loss results in an increase in NO levels at the wound edge. We have already shown that endothelial damage induces a long-lasting Ca2+ entry into surviving cells though connexin hemichannels (CxHcs) uncoupled from their counterparts on ruptured cells. The physiological outcome of injury-induced Ca2+ inflow is, however, unknown. Methods In this study, we sought to determine whether and how endothelial scraping induces NO production (NOP) in the endothelium of excised rat aorta by exploiting the NO-sensitive fluorochrome, DAF-FM diacetate and the Ca2+-sensitive fluorescent dye, Fura-2/AM. Results We demonstrated that injury-induced NOP at the lesion site is prevented in presence of the endothelial NO synthase inhibitor, L-NAME, and in absence of extracellular Ca2+. Unlike ATP-dependent NO liberation, the NO response to injury is insensitive to BTP-2, which selectively blocks store-operated Ca2+ inflow. However, injury-induced NOP is significantly reduced by classic gap junction blockers, and by connexin mimetic peptides specifically targeting Cx37Hcs, Cx40HCs, and Cx43Hcs. Moreover, disruption of caveolar integrity prevents injury-elicited NO signaling, but not the accompanying Ca2+ response. Conclusions The data presented provide the first evidence that endothelial scraping stimulates NO synthesis at the wound edge, which might both exert an immediate anti-thrombotic and anti-inflammatory action and promote the subsequent re-endothelialization.
Collapse
|
28
|
Moccia F, Dragoni S, Cinelli M, Montagnani S, Amato B, Rosti V, Guerra G, Tanzi F. How to utilize Ca²⁺ signals to rejuvenate the repairative phenotype of senescent endothelial progenitor cells in elderly patients affected by cardiovascular diseases: a useful therapeutic support of surgical approach? BMC Surg 2013; 13 Suppl 2:S46. [PMID: 24267290 PMCID: PMC3851045 DOI: 10.1186/1471-2482-13-s2-s46] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Endothelial dysfunction or loss is the early event that leads to a host of severe cardiovascular diseases, such as atherosclerosis, hypertension, brain stroke, myocardial infarction, and peripheral artery disease. Ageing is regarded among the most detrimental risk factor for vascular endothelium and predisposes the subject to atheroscleorosis and inflammatory states even in absence of traditional comorbid conditions. Standard treatment to restore blood perfusion through stenotic arteries are surgical or endovascular revascularization. Unfortunately, ageing patients are not the most amenable candidates for such interventions, due to high operative risk or unfavourable vascular involvement. It has recently been suggested that the transplantation of autologous bone marrow-derived endothelial progenitor cells (EPCs) might constitute an alternative and viable therapeutic option for these individuals. Albeit pre-clinical studies demonstrated the feasibility of EPC-based therapy to recapitulate the diseased vasculature of young and healthy animals, clinical studies provided less impressive results in old ischemic human patients. One hurdle associated to this kind of approach is the senescence of autologous EPCs, which are less abundant in peripheral blood and display a reduced pro-angiogenic activity. Conversely, umbilical cord blood (UCB)-derived EPCs are more suitable for cellular therapeutics due to their higher frequency and sensitivity to growth factors, such as vascular endothelial growth factor (VEGF). An increase in intracellular Ca2+ concentration is central to EPC activation by VEGF. We have recently demonstrated that the Ca2+ signalling machinery driving the oscillatory Ca2+ response to this important growth factor is different in UCB-derived EPCs as compared to their peripheral counterparts. In particular, we focussed on the so-called endothelial colony forming cells (ECFCs), which are the only EPC population belonging to the endothelial lineage and able to form capillary-like structures in vitro and stably integrate with host vasculature in vivo. The present review provides a brief description of how exploiting the Ca2+ toolkit of juvenile EPCs to restore the repairative phenotype of senescent EPCs to enhance their regenerative outcome in therapeutic settings.
Collapse
|
29
|
Grote K, Sonnenschein K, Kapopara PR, Hillmer A, Grothusen C, Salguero G, Kotlarz D, Schuett H, Bavendiek U, Schieffer B. Toll-like receptor 2/6 agonist macrophage-activating lipopeptide-2 promotes reendothelialization and inhibits neointima formation after vascular injury. Arterioscler Thromb Vasc Biol 2013; 33:2097-104. [PMID: 23868938 DOI: 10.1161/atvbaha.113.301799] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Reendothelialization after vascular injury (ie, balloon angioplasty or stent implantation) is clinically extremely relevant to promote vascular healing. We here investigated the therapeutic potential of the toll-like receptor 2/6 agonist macrophage-activating lipopeptide (MALP)-2 on reendothelialization and neointima formation in a murine model of vascular injury. APPROACH AND RESULTS The left common carotid artery was electrically injured, and reendothelialization was quantified by Evans blue staining after 3 days. A single injection of MALP-2 (1 or 10 µg, IV) after vascular injury accelerated reendothelialization (P<0.001). Proliferation of endothelial cells at the wound margins determined by 5-ethynyl-2'-deoxyuridine incorporation was significantly higher in MALP-2-treated animals (P<0.05). Furthermore, wire injury-induced neointima formation of the left common carotid artery was completely prevented by a single injection of MALP-2 (10 µg, IV). In vitro, MALP-2 induced proliferation (BrdU incorporation) and closure of an artificial wound of endothelial cells (P<0.05) but not of smooth muscle cells. Protein array and ELISA analysis of isolated primary endothelial cells and ex vivo stimulated carotid segments revealed that MALP-2 stimulated the release of multiple growth factors and cytokines predominantly from endothelial cells. MALP-2 induced a strong activation of the mitogen-activated protein kinase cascade in endothelial cells, which was attenuated in smooth muscle cells. Furthermore, MALP-2 significantly enhanced circulating monocytes and hematopoietic progenitor cells. CONCLUSIONS The toll-like receptor 2/6 agonist MALP-2 promotes reendothelialization and inhibits neointima formation after experimental vascular injury via enhanced proliferation and migration of endothelial cells. Thus, MALP-2 represents a novel therapeutic option to accelerate reendothelialization after vascular injury.
Collapse
Affiliation(s)
- Karsten Grote
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Antagonistic effect of C19 on migration of vascular smooth muscle cells and intimal hyperplasia induced by chemokine-like factor 1. Mol Biol Rep 2012. [PMID: 23203409 DOI: 10.1007/s11033-012-2309-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A chemokine-like factor 1 (CKLF1) is a recently discovered chemokine with broad-spectrum biological functions in inflammation and autoimmune diseases. C19 as a CKLF1's C-terminal peptide has been reported to exert inhibitory effects in a variety of diseases. However, the roles of CKLF1 and C19 on vascular smooth muscle cell (VSMC) migration and neointima formation still remain elusive. The effects of CKLF1 and C19 on VSMC migration and neointimal formation were investigated in cultured VSMCs and balloon-injured rat carotid arteries based on techniques including adenovirus-induced CKLF1 overexpression, gel based perivascular administration of C19, Boyden chamber, scratch-wound assay, real-time PCR, western blot and immunohistochemical analysis. CKLF1 was noticed to accumulate preferentially in neointima after the injury and colocalize with VSMCs. Luminal delivery of CKLF1 adenovirus to arteries exacerbated intimal thickening while perivascular administration of C19 to injured arteries attenuated this problem. In cultured primary VSMCs, CKLF1 overexpression up-regulated VSMC migration, which was down-regulated by C19. These data suggest that CKLF1 has a pivotal role in intimal hyperplasia by mediating VSMC migration. C19 was demonstrated to inhibit CKLF1-mediatated chemotaxis and restenosis. Thus further studies on C19 may provide a new treatment perspective for atherosclerosis and post-angioplasty restenosis.
Collapse
|
31
|
Fougerat A, Smirnova NF, Gayral S, Malet N, Hirsch E, Wymann MP, Perret B, Martinez LO, Douillon M, Laffargue M. Key role of PI3Kγ in monocyte chemotactic protein-1-mediated amplification of PDGF-induced aortic smooth muscle cell migration. Br J Pharmacol 2012; 166:1643-53. [PMID: 22251152 DOI: 10.1111/j.1476-5381.2012.01866.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Vascular smooth muscle cell (SMC) migration within the arterial wall is a crucial event in atherogenesis and restenosis. Monocyte chemotactic protein-1/CC-chemokine receptor 2 (MCP-1/CCR2) signalling is involved in SMC migration processes but the molecular mechanisms have not been well characterized. We investigated the role of PI3Kγ in SMC migration induced by MCP-1. EXPERIMENTAL APPROACHES A pharmacological PI3Kγ inhibitor, adenovirus encoding inactive forms of PI3Kγ and genetic deletion of PI3Kγ were used to investigate PI3Kγ functions in the MCP-1 and platelet-derived growth factor (PDGF) signalling pathway and migration process in primary aortic SMC. KEY RESULTS The γ isoform of PI3K was shown to be the major signalling molecule mediating PKB phosphorylation in MCP-1-stimulated SMC. Using a PI3Kγ inhibitor and an adenovirus encoding a dominant negative form of PI3Kγ, we demonstrated that PI3Kγ is essential for SMC migration triggered by MCP-1. PDGF receptor stimulation induced MCP-1 mRNA and protein accumulation in SMCs. Blockade of the MCP-1/CCR2 pathway or pharmacological inhibition of PI3Kγ reduced PDGF-stimulated aortic SMC migration by 50%. Thus PDGF promotes an autocrine loop involving MCP-1/CCR2 signalling which is required for PDGF-mediated SMC migration. Furthermore, SMCs isolated from PI3Kγ-deficient mice (PI3Kγ(-/-)), or mice expressing an inactive PI3Kγ (PI3Kγ(KD/KD)), migrated less than control cells in response to MCP-1 and PDGF. CONCLUSIONS AND IMPLICATIONS PI3Kγ is essential for MCP-1-stimulated aortic SMC migration and amplifies cell migration induced by PDGF by an autocrine/paracrine loop involving MCP-1 secretion and CCR2 activation. PI3Kγ is a promising target for the treatment of aortic fibroproliferative pathologies.
Collapse
|
32
|
Chalmers S, Saunter C, Wilson C, Coats P, Girkin JM, McCarron JG. Mitochondrial motility and vascular smooth muscle proliferation. Arterioscler Thromb Vasc Biol 2012; 32:3000-11. [PMID: 23104850 DOI: 10.1161/atvbaha.112.255174] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Mitochondria are widely described as being highly dynamic and adaptable organelles, and their movement is thought to be vital for cell function. Yet, in various native cells, including those of heart and smooth muscle, mitochondria are stationary and rigidly structured. The significance of the differences in mitochondrial behavior to the physiological function of cells is unclear and was studied in single myocytes and intact resistance-sized cerebral arteries. We hypothesized that mitochondrial dynamics is controlled by the proliferative status of the cells. METHODS AND RESULTS High-speed fluorescence imaging of mitochondria in live vascular smooth muscle cells shows that the organelle undergoes significant reorganization as cells become proliferative. In nonproliferative cells, mitochondria are individual (≈ 2 μm by 0.5 μm), stationary, randomly dispersed, fixed structures. However, on entering the proliferative state, mitochondria take on a more diverse architecture and become small spheres, short rod-shaped structures, long filamentous entities, and networks. When cells proliferate, mitochondria also continuously move and change shape. In the intact pressurized resistance artery, mitochondria are largely immobile structures, except in a small number of cells in which motility occurred. When proliferation of smooth muscle was encouraged in the intact resistance artery, in organ culture, the majority of mitochondria became motile and the majority of smooth muscle cells contained moving mitochondria. Significantly, restriction of mitochondrial motility using the fission blocker mitochondrial division inhibitor prevented vascular smooth muscle proliferation in both single cells and the intact resistance artery. CONCLUSIONS These results show that mitochondria are adaptable and exist in intact tissue as both stationary and highly dynamic entities. This mitochondrial plasticity is an essential mechanism for the development of smooth muscle proliferation and therefore presents a novel therapeutic target against vascular disease.
Collapse
Affiliation(s)
- Susan Chalmers
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | | | | | | | | | | |
Collapse
|
33
|
The effect of endothelial progenitor cells on angiotensin II-induced proliferation of cultured rat vascular smooth muscle cells. J Cardiovasc Pharmacol 2012; 58:617-25. [PMID: 22146405 DOI: 10.1097/fjc.0b013e318230bb5f] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Previous studies have demonstrated that endothelial progenitor cells (EPCs) could delay the progress of vascular remodeling in blood vessel-proliferating diseases. The proliferation of vascular smooth muscle cells (VSMCs) is a pivotal factor in cardiovascular diseases. In this study, we investigated whether EPCs could inhibit the Angiotensin II (Ang II)-induced proliferation of VSMCs. The effect of early EPC-conditioned medium (E-EPC-CM), late EPCs-CM (L-EPC-CM), and HUVEC-CM on Ang II-induced proliferation of VSMCs was assessed by BrdU incorporation, total protein content, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, and flow cytometry. Reverse transcriptase-polymerase chain reaction and Western blot were performed to analyze the effect of different CMs on Ang II-induced phosphorylations of ERK, JNK, p38, and NF-κB subunit p65 and the expressions of c-myc and c-fos. E-EPC-CM, L-EPC-CM, and HUVEC-CM significantly inhibited the Ang II-induced DNA synthesis, total protein expression, cell survival, and cell cycle progress of VSMCs. Furthermore, E-EPC-CM significantly inhibited the Ang II-induced phosphorylation of ERK, JNK, p38, and p65 (nuclear translocation of p65) and the expressions of c-myc and c-fos. Taken together, these data suggested that EPCs may delay the progress of vascular remodeling in blood vessel-proliferating diseases by inhibiting Ang II-induced proliferation of VSMCs through inactivating MAPKs and NF-κB signaling pathways and by reducing the expressions of c-myc and c-fos.
Collapse
|
34
|
Song Z, Jin R, Yu S, Nanda A, Granger DN, Li G. Crucial role of CD40 signaling in vascular wall cells in neointimal formation and vascular remodeling after vascular interventions. Arterioscler Thromb Vasc Biol 2012; 32:50-64. [PMID: 21998133 PMCID: PMC3241889 DOI: 10.1161/atvbaha.111.238329] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE It has been shown that CD40-TRAF6 axis in leukocytes plays a significant role in neointimal formation after carotid ligation. Because CD40 and TRAF6 are expressed not only in leukocytes but also in vascular cells, we examined the role of CD40 contributed by vascular wall cells in neointimal formation after carotid ligation in an atherogenic environment. METHODS AND RESULTS Both CD40 and TRAF6 in medial smooth muscle cells (SMCs) was upregulated significantly at 3 days and more prominently at 7 days after injury in wildtype mice, but the TRAF6 upregulation was abolished in CD40(-/-) mice. In vitro, TRAF6 expression was induced by cytokines (tumor necrosis factor -α, interleukin-1β) via a NF-κB-dependent manner in wildtype SMCs, but this induction was blocked in CD40-deficient SMCs. Bone marrow chimeras revealed a comparable reduction in neointimal formation and lumen stenosis in mice lacking either vascular wall- or bone marrow-associated CD40. Lacking vascular wall-associated CD40 resulted in a significant reduction in monocyte/macrophage accumulation, NF-κB activation, and multiple proinflammatory mediators (ICAM-1, VCAM-1, MCP-1, MMP-9, tissue factor). In vitro data confirmed that CD40 deficiency or TRAF6 knockdown suppressed CD40L-induced proinflammatory phenotype of SMCs by inhibition of NF-κB activation. Moreover, both in vivo and in vitro data showed that CD40 deficiency prevented injury-induced SMC apoptosis but did not affect SMC proliferation and migration. CONCLUSIONS CD40 signaling through TRAF6 in vascular SMCs seems to be centrally involved in neointimal formation in a NF-κB-dependent manner. Modulating CD40 signaling on local vascular wall may become a new therapeutic target against vascular restenosis.
Collapse
Affiliation(s)
- Zifang Song
- Department of Neurosurgery, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA
| | | | | | | | | | | |
Collapse
|
35
|
Berglund LM, Kotova O, Osmark P, Grufman H, Xing C, Lydrup ML, Goncalves I, Autieri MV, Gomez MF. NFAT regulates the expression of AIF-1 and IRT-1: yin and yang splice variants of neointima formation and atherosclerosis. Cardiovasc Res 2011; 93:414-23. [PMID: 22116621 DOI: 10.1093/cvr/cvr309] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Alternative transcription and splicing of the allograft inflammatory factor-1 (AIF-1) gene results in the expression of two different proteins: AIF-1 and interferon responsive transcript-1 (IRT-1). Here, we explore the impact of AIF-1 and IRT-1 on vascular smooth muscle cell (VSMC) activation and neointima formation, the mechanisms underlying their alternative splicing, and associations of AIF-1 and IRT-1 mRNA with parameters defining human atherosclerotic plaque phenotype. METHODS AND RESULTS Translation of AIF-1 and IRT-1 results in different products with contrasting cellular distribution and functions. Overexpression of AIF-1 stimulates migration and proliferation of human VSMCs, whereas IRT-1 exerts opposite effects. Adenoviral infection of angioplasty-injured rat carotid arteries with AdAIF-1 exacerbates intima hyperplasia, whereas infection with AdIRT-1 reduces neointima. Expression of these variants is modulated by changes in nuclear factor of activated T-cells (NFAT) activity. Pharmacological inhibition of NFAT or targeting of NFATc3 with small interfering RNA (siRNA) lowers the AIF-1/IRT-1 ratio and favours an anti-proliferative outcome. NFAT acts as a repressor on the IRT-1 transcriptional start site, which is also sensitive to interferon-γ stimulation. Expression of AIF-1 mRNA in human carotid plaques associates with less extracellular matrix and a more pro-inflammatory plaque and plasma profile, features that may predispose to plaque rupture. In contrast, expression of IRT-1 mRNA associates with a less aggressive phenotype and less VSMCs at the most stenotic region of the plaque. CONCLUSION Inhibition of NFAT signalling, by shifting the AIF-1/IRT-1 ratio, may be an attractive target to regulate the VSMC response to injury and manipulate plaque stability in atherosclerosis.
Collapse
Affiliation(s)
- Lisa M Berglund
- Department of Clinical Sciences, Lund University, CRC, Entrance 72, Building 91, Level 12, 205 02 Malmö, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fang L, Chen MF, Xiao ZL, Liu Y, Yu GL, Chen XB, Xie XM. Calcitonin gene-related peptide released from endothelial progenitor cells inhibits the proliferation of rat vascular smooth muscle cells induced by angiotensin II. Mol Cell Biochem 2011; 355:99-108. [PMID: 21603886 DOI: 10.1007/s11010-011-0843-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Accepted: 04/20/2011] [Indexed: 02/07/2023]
Abstract
We have recently demonstrated that endothelial progenitor cells (EPCs) inhibit AngII-induced proliferation of vascular smooth muscle cells (VSMCs) by inactivating MAPKs and NF-κB signaling pathway and reducing expression of oncogene c-myc and c-fos. The inhibitory effect of EPCs on VSMCs is associated with paracrine mechanism. However, the potential mechanism of EPCs on the regulation of AngII-induced proliferation of VSMCs was unknown. Calcitonin gene-related peptide (CGRP) could inhibit AngII-induced proliferation and transformation of VSMCs. However, it has not been known whether CGRP released from EPCs is a potential regulator in regulation of AngII-induced proliferation of VSMCs. Early endothelial progenitor cell-conditioned medium(E-EPC-CM) was pre-incubated with functional blocking antibodies against CGRP for 1 h or VSMCs was preteated with CGRP(837)(CGRP receptor antagonist) for 1 h before VSMCs were pretreated with CM for 30 min. DNA synthesis ability, total protein levels, cell survival, signal transduction, and expressions of c-myc and c-fos of VSMCs induced by AngII (10(-6)mol/l) were detected to assess the role of CGRP in AngII-induced proliferation of VSMCs. E-EPC-CM could significantly inhibit AngII-induced DNA synthesis ability, total protein levels, cell survival, phosphorylation of ERK, JNK, p38, p65, and expressions of c-myc and c-fos compared with the control group(P < 0.05). However, Pretreatment with anti-CGRP antibody and CGRP(837) could significantly weaken the inhibitory effect of E-EPC-CM on proliferation of VSMCs induced by AngII (P < 0.05). EPCs exert anti-proliferative effects on VSMCs mediated by the release of CGRP.
Collapse
Affiliation(s)
- Li Fang
- Department of Geriatric Cardiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Triptolide inhibits rat vascular smooth muscle cell proliferation and cell cycle progression via attenuation of ERK1/2 and Rb phosphorylation. Exp Mol Pathol 2011; 90:137-42. [DOI: 10.1016/j.yexmp.2010.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 12/06/2010] [Indexed: 01/31/2023]
|
38
|
Weakley SM, Wang X, Mu H, Lü J, Lin PH, Yao Q, Chen C. Ginkgolide A-gold nanoparticles inhibit vascular smooth muscle proliferation and migration in vitro and reduce neointimal hyperplasia in a mouse model. J Surg Res 2011; 171:31-9. [PMID: 21571322 DOI: 10.1016/j.jss.2011.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 02/17/2011] [Accepted: 03/04/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Neointimal formation is mediated by phenotypic changes in vascular smooth muscle cells (SMC) and is an important mediator of restenosis following arterial reconstruction. We conjugated antioxidant ginkgolide A (GA) to gold nanoparticles (GNP) to determine the effect of GA delivery on neointimal formation. MATERIALS AND METHODS GA was conjugated to 80 nm GNP in an overnight incubation. Mouse P53LMAC01 vascular SMC were treated with various doses of GA-GNP, GA alone, GNP alone, and no treatment control. Cell proliferation and migration were analyzed, and superoxide anion levels and the phosphorylation status of ERK1/2 were determined. Mice underwent ligation of the common carotid artery along with local treatment with GNP (control) or GA-GNP. The carotid artery was harvested and subjected to immunohistochemical analysis. RESULTS GA-GNP treatment significantly inhibited SMC proliferation and migration in vitro in comparison to GNP treatment alone, and the effect persisted for up to 72 h after treatment. Treatment with GA-GNP also reduced superoxide anion levels in vitro. PDGF-BB substantially induced ERK1/2 phosphorylation in GNP control cells; this PDGE-BB induced ERK1/2 phosphorylation was significantly inhibited in GA-GNP-treated cells compared with GNP only. GA-GNP significantly reduced neointimal hyperplasia after injury in mice, and proliferating cell nuclear antigen (PCNA) staining was reduced substantially in the arteries of mice treated with GA-GNP. CONCLUSIONS GA-GNP reduce vascular SMC proliferation and migration in vitro through reduced activation of ERK1/2. Local treatment with GA-GNP in areas of arterial injury reduced neointimal hyperplasia and subsequent stenosis.
Collapse
Affiliation(s)
- Sarah M Weakley
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. 20-HETE and EETs are generated in various cell types in the brain and cerebral blood vessels, and contribute significantly to cerebral blood flow autoregulation and the coupling of regional brain blood flow to neuronal activity (neurovascular coupling). Investigations are beginning to unravel the molecular and cellular mechanisms by which these CYP eicosanoids regulate cerebral vascular function and the changes that occur in pathological states. Intriguingly, 20-HETE and the soluble epoxide hydrolase (sEH) enzyme that regulates EET levels have been explored as molecular therapeutic targets for cerebral vascular diseases. Inhibition of 20-HETE, or increasing EET levels by inhibiting the sEH enzyme, decreases cerebral damage following stroke. The improved outcome following cerebral ischaemia is a consequence of improving cerebral vascular structure or function and protecting neurons from cell death. Thus, the CYP eicosanoids are key regulators of cerebral vascular function and novel therapeutic targets for cardiovascular diseases and neurological disorders.
Collapse
|
40
|
Chieng-Yane P, Bocquet A, Létienne R, Bourbon T, Sablayrolles S, Perez M, Hatem SN, Lompré AM, Le Grand B, David-Dufilho M. Protease-activated receptor-1 antagonist F 16618 reduces arterial restenosis by down-regulation of tumor necrosis factor α and matrix metalloproteinase 7 expression, migration, and proliferation of vascular smooth muscle cells. J Pharmacol Exp Ther 2011; 336:643-51. [PMID: 21139058 DOI: 10.1124/jpet.110.175182] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Wound healing after angioplasty or stenting is associated with increased production of thrombin and the activation of protease-activated receptor 1 (PAR1). The aim of the present study was to examine the effects of a new selective PAR1 antagonist, 2-[5-oxo-5-(4-pyridin-2-ylpiperazin-1-yl)-penta-1,3-dienyl]-benzonitrile (F 16618), in restenosis and vascular smooth muscle cell (SMC) proliferation and migration using both in vivo and in vitro approaches. Daily oral administration of F 16618 inhibited the restenosis induced by balloon angioplasty on rat carotid artery in a dose-dependent manner. Furthermore, single intravenous administration of F 16618 during the angioplasty procedure was sufficient to protect the carotid artery against restenosis. In vitro, F 16618 inhibited the growth of human aortic SMCs in a concentration-dependent manner with maximal effects at 10 μM. At that concentration, F 16618 also prevented thrombin-mediated SMC migration. In vivo, oral and intravenous F 16618 treatments reduced by 30 and 50% the expression of the inflammatory cytokine tumor necrosis factor α (TNFα) 24 h after angioplasty. However, only acute intravenous administration prevented the induction of matrix metalloproteinase 7 expression. In contrast, F 16618 treatments had no effect on early SMC de-differentiation and transcription of monocyte chemoattractant protein-1 and interleukin-6 and late re-endothelialization of injured arteries. Furthermore, F 16618 compensated for the carotid endothelium loss by inhibiting PAR1-mediated contraction. Altogether, these data demonstrate that PAR1 antagonists such as F 16618 are a highly effective treatment of restenosis after vascular injury, by inhibition of TNFα, matrix metalloproteinase 7, and SMC migration and proliferation in addition to an antithrombotic effect.
Collapse
MESH Headings
- Animals
- Carotid Stenosis/metabolism
- Carotid Stenosis/prevention & control
- Cell Movement/drug effects
- Cell Movement/physiology
- Cell Proliferation/drug effects
- Cells, Cultured
- Coronary Restenosis/drug therapy
- Coronary Restenosis/metabolism
- Down-Regulation/drug effects
- Down-Regulation/physiology
- Gene Expression Regulation, Enzymologic
- Humans
- Male
- Matrix Metalloproteinase 7/biosynthesis
- Matrix Metalloproteinase Inhibitors
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Pyridines/pharmacology
- Pyridines/therapeutic use
- Rats
- Rats, Sprague-Dawley
- Receptor, PAR-1/physiology
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/metabolism
- Platelet Aggregation Inhibitors
Collapse
|
41
|
Matrix metalloproteinases modulated by protein kinase Cε mediate resistin-induced migration of human coronary artery smooth muscle cells. J Vasc Surg 2011; 53:1044-51. [PMID: 21277149 DOI: 10.1016/j.jvs.2010.10.117] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 10/20/2010] [Accepted: 10/24/2010] [Indexed: 12/19/2022]
Abstract
BACKGROUND Emerging evidence showed that resistin induces vascular smooth muscle cell (VSMC) migration, a critical step in initiating vascular restenosis. Adhesion molecule expression and cytoskeletal rearrangement have been observed in this progress. Given that matrix metalloproteinases (MMPs) also regulate cell migration, we hypothesized that MMPs may mediate resistin-induced VSMC migration. METHODS Human VSMCs were treated with recombinant human resistin at physiologic (10 ng/mL) and pathologic (40 ng/mL) concentrations for 24 hours. Cell migration was determined by the Boyden chamber assay. MMP and tissue inhibitor metalloproteinase (TIMP) mRNA and protein levels were measured with real-time PCR and ELISA. MMP enzymatic activity was measured by zymography. In another experiment, neutralizing antibodies against MMP-2 and MMP-9 were coincubated with resistin in cultured VSMCs. The regulation of MMP by protein kinase C (PKC) was determined by εV1-2, a selective PKCε inhibitor. RESULTS Resistin-induced smooth muscle cell (SMC) migration was confirmed by the Boyden chamber assay. Forty nanograms/milliliter resistin increased SMC migration by 3.7 fold. Additionally, resistin stimulated MMP-2 and -MMP9 mRNA and protein expressions. In contrast, the TIMP-1 and TIMP-2 mRNA levels were inhibited by resistin. Neutralizing antibodies against MMP-2 and MMP-9 effectively reversed VSMC migration. Furthermore, resistin activated PKCε, but selective PKCε inhibitor suppressed resistin-induced MMP expression, activity, and cell migration. CONCLUSIONS Our study confirmed that resistin increased vascular smooth muscle cell migration in vitro. In terms of mechanism, resistin-stimulated cell migration was associated with increased MMP expression, which was dependent on PKCε activation.
Collapse
|
42
|
Ding RQ, Tsao J, Chai H, Mochly-Rosen D, Zhou W. Therapeutic potential for protein kinase C inhibitor in vascular restenosis. J Cardiovasc Pharmacol Ther 2010; 16:160-7. [PMID: 21183728 DOI: 10.1177/1074248410382106] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vascular restenosis, an overreaction of biological response to injury, is initialized by thrombosis and inflammation. This response is characterized by increased smooth muscle cell migration and proliferation. Available pharmacological treatments include anticoagulants, antiplatelet agents, immunosuppressants, and antiproliferation agents. Protein kinase C (PKC), a large family of serine/threonine kinases, has been shown to participate in various pathological stages of restenosis. Consequently, PKC inhibitors are expected to exert a wide range of pharmacological activities therapeutically beneficial for restenosis. In this review, the roles of PKC isozymes in platelets, leukocytes, endothelial cells, and smooth muscle cells are discussed, with emphasis given to smooth muscle cells. We will describe cellular and animal studies assessing prevention of restenosis with PKC inhibitors, particularly targeting -α, -β, -δ, and -ζ isozymes. The delivery strategy, efficacy, and safety of such PKC regulators will also be discussed.
Collapse
Affiliation(s)
- Richard Qinxue Ding
- Division of Vascular and Endovascular Surgery, Department of Surgery, Stanford University, Stanford, CA 94350, USA
| | | | | | | | | |
Collapse
|
43
|
Hong SJ, Kim ST, Kim TJ, Kim EO, Ahn CM, Park JH, Kim JS, Lee KM, Lim DS. Cellular and Molecular Changes Associated With Inhibitory Effect of Pioglitazone on Neointimal Growth in Patients With Type 2 Diabetes After Zotarolimus-Eluting Stent Implantation. Arterioscler Thromb Vasc Biol 2010; 30:2655-65. [DOI: 10.1161/atvbaha.110.212670] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Soon Jun Hong
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Sung Tae Kim
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Tae-Jin Kim
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Eun-Ok Kim
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Chul-Min Ahn
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Jae Hyoung Park
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Je Sang Kim
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Kyung-Mi Lee
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| | - Do-Sun Lim
- From the Department of Cardiology (S.J.H., C.-M.A., J.H.P., J.S.K., D.-S.L.), Korea University Anam Hospital, Seoul, Korea; and the Department of Biochemistry and Molecular Biology (S.T.K., T.-J.K., E.O.K., K.-M.L.), Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
Köhler R, Kaistha BP, Wulff H. Vascular KCa-channels as therapeutic targets in hypertension and restenosis disease. Expert Opin Ther Targets 2010; 14:143-55. [PMID: 20055714 DOI: 10.1517/14728220903540257] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
IMPORTANCE OF THE FIELD Cardiovascular disease is a leading cause of death in modern societies. Hyperpolarizing Ca(2+)-activated K(+) channels (K(Ca)) are important membrane proteins in the control of arterial tone and pathological vascular remodelling and thus could serve as new drug targets. AREAS COVERED IN THIS REVIEW We summarize recent advances in the field of vascular K(Ca) and their roles in cardiovascular pathologies such as hypertension and restenosis disease and draw attention to novel small-molecule channel modulators and their possible therapeutic utility. This review focuses on literature from the last four to five years. WHAT THE READER WILL GAIN Pharmacological opening of endothelial KCa3.1/KCa2.3 channels stimulates endothelium-derived-hyperpolarizing-factor-mediated arteriolar dilation and lowers blood pressure. Inhibition of smooth muscle KCa3.1 channels has beneficial effects in restenosis disease and atherosclerosis. We consider the therapeutic potential of KCa3.1/KCa2.3 openers as novel endothelium-specific antihypertensive drugs as well as of KCa3.1-blockers for the treatment of pathological vascular remodelling and discuss advantages and disadvantages of the pharmacotherapeutic approaches. TAKE HOME MESSAGE Pharmacological manipulation of vascular K(Ca) channels by novel small-molecule modulators offers new venues for alternative treatments of hypertension, restenosis and atherosclerosis. Additional efforts are required to optimize these compounds and to validate them as cardiovascular-protective drugs.
Collapse
Affiliation(s)
- Ralf Köhler
- Institute of Medical Biology, Depatrment of Physiology, SDU, J.B. Winsløwsvej 21 3., 5000 Odense C, Denmark.
| | | | | |
Collapse
|
45
|
Mrabat H, Garg HG, Hales CA. Growth inhibition of bovine pulmonary artery smooth muscle cells following long-term heparin treatment. J Cell Physiol 2009; 221:603-8. [PMID: 19653229 PMCID: PMC2755647 DOI: 10.1002/jcp.21891] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Heparin (HP) inhibits pulmonary artery smooth muscle cell (PASMC) growth in vitro and vascular remodeling in vivo. Bârzu et al. (1994) suggested that the antiproliferative effect of HP on rat aortic smooth muscle cell in vitro diminishes with prolonged exposure to heparin. We exposed cultured bovine PASMC (BPASMC) to prolonged pretreatment with 20 microg/ml of 0-hexanoylated HP from passages 3 to13 and compared them to control (no pretreatment) cultures of identical passages. The pretreated BPASMC and control groups were growth arrested for 48 h, followed by treatment of 0-hexanoylated HP at different doses. On day 5, the growth inhibition of BPASMC was determined. The percent inhibition by 1 microg/ml of 0-hexanoylated HP was 46 +/- 14% versus 62 +/- 13%, for control and pretreated BPASMC, respectively. At 10 microg/ml the inhibition was 62 +/- 7% versus 84 +/- 6%. For 100 microg/ml the inhibition increased to 92 +/- 5% versus 100% and at 200 microg/ml the inhibition was 95 +/- 3% versus 100%. BPASMC (with or without preexposure to 0-hexanoylated HP), at passage 13, were sensitive to the growth inhibitory effect of 0-hexanoylated HP with no significant difference among the groups (95 +/- 3% inhibition vs. 100% for pretreated BPASMC). We found that 0-hexanoylated HP-induced necrosis as shown by flow cytometry and only minor apoptosis. Caspase-3 and PARP detection was insignificant between the groups. In summary, no cell subpopulation at long-term treatment exhibited resistance to 0-hexanoylated HP. The HP antiproliferative effect on SMC is potentially important in defining new approaches to the treatment of the remodeled vasculature of pulmonary hypertension. Liss, Inc.
Collapse
Affiliation(s)
- Hicham Mrabat
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | |
Collapse
|
46
|
Csányi G, Taylor WR, Pagano PJ. NOX and inflammation in the vascular adventitia. Free Radic Biol Med 2009; 47:1254-66. [PMID: 19628034 PMCID: PMC3061339 DOI: 10.1016/j.freeradbiomed.2009.07.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 07/10/2009] [Accepted: 07/14/2009] [Indexed: 02/07/2023]
Abstract
Vascular inflammation has traditionally been thought to be initiated at the luminal surface and progress through the media toward the adventitial layer. In recent years, however, evidence has emerged suggesting that the vascular adventitia is activated early in a variety of cardiovascular diseases and that it plays an important role in the initiation and progression of vascular inflammation. Adventitial fibroblasts have been shown to produce substantial amounts of NAD(P)H oxidase-derived reactive oxygen species (ROS) in response to vascular injury. Additionally, inflammatory cytokines, lipids, and various hormones, implicated in fibroblast proliferation and migration, lead to recruitment of inflammatory cells to the adventitial layer and impairment of endothelium-dependent relaxation. Early in the development of vascular disease, there is clear evidence for progression toward a denser vasa vasorum which delivers oxygen and nutrients to an increasingly hypoxic and nutrient-deficient media. This expanded vascularization appears to provide enhanced delivery of inflammatory cells to the adventitia and outer media. Combined adventitial fibroblast and inflammatory cell-derived ROS therefore are expected to synergize their local effect on adventitial parenchymal cells, leading to further cytokine release and a feed-forward propagation of adventitial ROS production. In fact, data from our laboratory and others suggest a broader paracrine positive feedback role for adventitia-derived ROS in medial smooth muscle cell hypertrophy and neointimal hyperplasia. A likely candidate responsible for the adventitia-derived paracrine signaling across the vessel wall is the superoxide anion metabolite hydrogen peroxide, which is highly stable, cell permeant, and capable of activating downstream signaling mechanisms in smooth muscle cells, leading to phenotypic modulation of smooth muscle cells. This review addresses the role of adventitial NAD(P)H oxidase-derived ROS from a nontraditional, perivascular vantage of promoting vascular inflammation and will discuss how ROS derived from adventitial NAD(P)H oxidases may be a catalyst for vascular remodeling and dysfunction.
Collapse
Affiliation(s)
- Gábor Csányi
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - W. Robert Taylor
- Departments of Medicine and Biomedical Engineering, Emory University and the Atlanta VA Medical Center, Atlanta, GA
| | - Patrick J. Pagano
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
47
|
Song M, Yu X, Cui X, Zhu G, Zhao G, Chen J, Huang L. Blockade of Connexin 43 Hemichannels Reduces Neointima Formation After Vascular Injury by Inhibiting Proliferation and Phenotypic Modulation of Smooth Muscle Cells. Exp Biol Med (Maywood) 2009; 234:1192-200. [PMID: 19596827 DOI: 10.3181/0902-rm-80] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Connexins 43 (Cx43) plays a key role in neointimal formation after vascular injury, but the mechanism still needs to be further explored. We hypothesized that the gap junction-dependent function of Cx43 to mediate intercellular communication has a crucial role in the development and progression of vascular diseases. The effect of intercellular communication mediated by Cx43 hemichannels on neointimal formation after vascular injury was investigated. Cx43 was overexpressed or knockdown in rat vascular smooth muscle cell (SMC) by transfection pcDNA-Cx43 plasmid or small interfering RNA (siRNA) against Cx43 (siCx43). SMC proliferation and marker genes expression after Cx43 alteration and blockade of the Cx43 hemichannel were analyzed by 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay and RT-PCR. The effect of carbenoxolone on neointimal formation was investigated in carotid artery injured rat model. We demonstrated that overexpression of Cx43 promoted SMC proliferation, meanwhile, mRNA expression level of smooth muscle α-actin and calponin, which were important markers of SMC in a contractile state, were down-regulated in smooth muscle. Knockdown of Cx43 inhibited SMC proliferation but increased SMC marker genes expression level. Carbenoxolone (50 μM) improved SMC contractile differentiation and inhibited its proliferation. Our data showed that carbenoxolone reduced neointimal formation after carotid artery injury. In summary, blockade of intercellular communication via Cx43 hemichannels reduces neointimal formation after vascular injury by inhibiting proliferation and phenotypic modulation of SMCs.
Collapse
MESH Headings
- Animals
- Carbenoxolone/pharmacology
- Carotid Artery Injuries/metabolism
- Cell Proliferation/drug effects
- Coloring Agents/metabolism
- Connexin 43/genetics
- Connexin 43/metabolism
- Endothelium, Vascular/metabolism
- Fluorescence Recovery After Photobleaching
- Fluorescent Antibody Technique, Indirect
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Peptides/pharmacology
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Tetrazolium Salts/metabolism
- Thiazoles/metabolism
- Transfection
- Tunica Intima/drug effects
- Tunica Intima/metabolism
Collapse
Affiliation(s)
- MingBao Song
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
48
|
Kusch A, Tkachuk S, Tkachuk N, Patecki M, Park JK, Dietz R, Haller H, Dumler I. The tight junction protein ZO-2 mediates proliferation of vascular smooth muscle cells via regulation of Stat1. Cardiovasc Res 2009; 83:115-22. [PMID: 19380416 DOI: 10.1093/cvr/cvp117] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Recent evidence suggests that the zonula occludens protein 2 (ZO-2) might have additional cellular functions, beyond regulation of paracellular permeability of epithelial and endothelial cells. Deregulation of ZO-2 in response to ischaemia, hypertensive stress, and vascular injury implies its involvement in cardiovascular disorders, most likely via regulating the functional behaviour of vascular smooth muscle cells (VSMC). However, a role of ZO-2 in VSMC biology has yet to be established. Our study was designed to understand the specific functions of ZO-2 in human VSMC. METHODS AND RESULTS The expression of ZO-2 and Stat1 upon vascular injury was studied using ex vivo organ culture of coronary arteries combined with immunohistochemistry. ZO-2 silencing in human primary VSMC was achieved by means of lentiviral gene transfer. Cell proliferation was assessed by analysing DNA synthesis and by cell counting. Stat1 expression was examined using immunoblotting, immunocytochemistry, TaqMan, and fluorescence activated cell sorting (FACS) analysis. Functional relevance of Stat1 up-regulation was studied using a Stat1 promoter-luciferase reporter assay and intracellular microinjections of a Stat1 specific antibody. ZO-2 was highly expressed in the media and neointima of dilated but not of control arteries, whereas expression of the transcription factor Stat1 was inversely regulated upon injury. Analysis of VSMC with down-regulated ZO-2 revealed increased expression of Stat1 in these cells, whereas Stat1 phosphorylation was not affected. Stat1 up-regulation in VSMC with ZO-2 silencing resulted in a coordinate activation of Stat1-specific genes and consequently led to inhibition of cell proliferation. This effect was restored by microinjection of a Stat1 neutralising antibody. CONCLUSION Our data suggest that the tight junction protein ZO-2 is involved in regulation of VSMC growth control upon vascular injury that is mediated by the transcription factor Stat1. Our findings point to a novel function of ZO-2 in VSMC and implicate ZO-2 as a novel important molecular target in pathological states of vascular remodelling in cardiovascular diseases.
Collapse
Affiliation(s)
- Angelika Kusch
- Medical Faculty of the Charité, Franz Volhard Klinik/Experimental and Clinical Research Center-ECRC, Max Delbrück Center, Berlin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lee MY, San Martin A, Mehta PK, Dikalova AE, Garrido AM, Datla SR, Lyons E, Krause KH, Banfi B, Lambeth JD, Lassègue B, Griendling KK. Mechanisms of vascular smooth muscle NADPH oxidase 1 (Nox1) contribution to injury-induced neointimal formation. Arterioscler Thromb Vasc Biol 2009; 29:480-7. [PMID: 19150879 DOI: 10.1161/atvbaha.108.181925] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Vascular NADPH oxidases (Noxes) have been implicated in cardiovascular diseases; however, the importance of individual Nox homologues remains unclear. Here, the role of the vascular smooth muscle cell (VSMC) Nox1 in neointima formation was studied using genetically modified animal models. METHODS AND RESULTS Wire injury-induced neointima formation in the femoral artery, along with proliferation and apoptosis, was reduced in Nox1(y/-) mice, but there was little difference in Tg(SMCnox1) mice compared with wild-type (WT) mice. Proliferation and migration were reduced in cultured Nox1(y/-) VSMCs and increased in Tg(SMCnox1) cells. Tg(SMCnox1) cells exhibited increased fibronectin secretion, but neither collagen I production nor cell adhesion was affected by alteration of Nox1. Using antibody microarray and Western blotting analysis, increased cofilin phosphorylation and mDia1 expression and decreased PAK1 expression were detected in Nox1(y/-) cells. Overexpression of S3A, a constitutively active cofilin mutant, partially recovered reduced migration of Nox1(y/-) cells, suggesting that reduction in cofilin activity contributes to impaired migration of Nox1(y/-) VSMCs. CONCLUSIONS These results indicate that Nox1 plays a critical role in neointima formation by mediating VSMC migration, proliferation, and extracellular matrix production, and that cofilin is a major effector of Nox1-mediated migration. Inhibition of Nox1 may be an efficient strategy to suppress neointimal formation.
Collapse
Affiliation(s)
- Moo Yeol Lee
- Emory University, Department of Medicine, Division of Cardiology, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hu T, Luan R, Zhang H, Lau WB, Wang Q, Zhang Y, Wang HC, Tao L. Hydrogen peroxide enhances osteopontin expression and matrix metalloproteinase activity in aortic vascular smooth muscle cells. Clin Exp Pharmacol Physiol 2008; 36:626-30. [PMID: 19076167 DOI: 10.1111/j.1440-1681.2008.05124.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Restenosis after percutaneous coronary intervention (PCI) is a major clinical complication. However, the underlying mechanisms remain poorly understood. The present aim of the present study was to test the hypothesis that reactive oxygen species (ROS) enhance osteopontin (OPN) expression and increase matrix metalloproteinase (MMP)-2 activity (two major factors that contribute to restenosis) in aortic vascular smooth muscle cells (VSMC), thus facilitating restenosis. 2. Primary cultured rat aortic VSMC were exposed to different concentrations (10, 50 and 100 micromol/L) of H(2)O(2). The expression of OPN mRNA and protein was determined by reverse transcription-polymerase chain reaction and Western blotting, respectively. The activity of MMP-2 was determined by gelatin zymography. 3. The expression of OPN mRNA and protein in VSMC was enhanced by H(2)O(2) in a dose-dependent manner. In addition, H(2)O(2) at all concentrations tested (which are comparable to those seen in diabetic vascular tissues) significantly increased MMP-2 activity in VSMC. 4. Because vascular ROS production is significantly increased in patients with ischaemic disease and OPN and MMP-2 have been shown to play critical role in restenosis, the results of the present study strongly suggest that a ROS-initiated and OPN- and MMP-2-mediated signalling pathway may play an important role in accelerated restenosis after PCI in patients with ischaemic disease. Therefore, the H(2)O(2)-OPN/MMP-2 system may be a new therapeutic target in reducing restenosis in patients undergoing PCI.
Collapse
Affiliation(s)
- Tao Hu
- Deparrment of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | |
Collapse
|