1
|
Lei J, Zheng F, Chen L, Zhang R, Yang Y, Yin Z, Luo L. Gstp1 negatively regulates blood pressure in hypertensive rat via promoting APLNR ubiquitination degradation mediated by Nedd4. Clin Sci (Lond) 2024; 138:883-900. [PMID: 38959295 DOI: 10.1042/cs20241113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/05/2024]
Abstract
Hypertension is a leading risk factor for disease burden worldwide. Vascular contraction and remodeling contribute to the development of hypertension. Glutathione S-transferase P1 (Gstp1) plays several critical roles in both normal and neoplastic cells. In this study, we investigated the effect of Gstp1 on hypertension as well as on vascular smooth muscle cell (VSMC) contraction and phenotypic switching. We identified the higher level of Gstp1 in arteries and VSMCs from hypertensive rats compared with normotensive rats for the first time. We then developed Adeno-associated virus 9 (AAV9) mediated Gstp1 down-regulation and overexpression in rats and measured rat blood pressure by using the tail-cuff and the carotid catheter method. We found that the blood pressure of spontaneously hypertensive rats (SHR) rose significantly with Gstp1 down-regulation and reduced apparently after Gstp1 overexpression. Similar results were obtained from the observations of 2-kidney-1-clip renovascular (2K1C) hypertensive rats. Gstp1 did not influence blood pressure of normotensive Wistar-Kyoto (WKY) rats and Sprague-Dawley (SD) rats. Further in vitro study indicated that Gstp1 knockdown in SHR-VSMCs promoted cell proliferation, migration, dedifferentiation and contraction, while Gstp1 overexpression showed opposite effects. Results from bioinformatic analysis showed that the Apelin/APLNR system was involved in the effect of Gstp1 on SHR-VSMCs. The rise in blood pressure of SHR induced by Gstp1 knockdown could be reversed by APLNR antagonist F13A. We further found that Gstp1 enhanced the association between APLNR and Nedd4 E3 ubiquitin ligases to induce APLNR ubiquitination degradation. Thus, in the present study, we discovered a novel anti-hypertensive role of Gstp1 in hypertensive rats and provided the experimental basis for designing an effective anti-hypertensive therapeutic strategy.
Collapse
Affiliation(s)
- Jianzhen Lei
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Fen Zheng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Luyao Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Ruyi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yang Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu 210046, China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| |
Collapse
|
2
|
Zhao Z, Yu H, Nie M, Li X, Li M. Remote Ischemic Preconditioning Before Drug-Coated Balloon Implantation can Improve the Long-Term Prognosis of Patients with CAD. Rev Cardiovasc Med 2024; 25:116. [PMID: 39076569 PMCID: PMC11264023 DOI: 10.31083/j.rcm2504116] [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: 10/15/2023] [Revised: 11/30/2023] [Accepted: 12/08/2023] [Indexed: 07/31/2024] Open
Abstract
Background Drug-coated balloons (DCBs) have become increasingly vital to percutaneous coronary intervention, offering many advantages. However, a significant challenge is that many patients are intolerant to the myocardial ischemia caused by DCB dilation. Remote ischemic preconditioning (RIPC) is known to enhance heart's tolerance to ischemia and hypoxia. This study investigated whether preoperative RIPC could extend the tolerated DCB inflation time and improve the long-term prognosis of patients with coronary artery disease (CAD). Methods A total of 653 patients with CAD were recruited and randomized into a RIPC group (n = 323) and a control (n = 330) group. The RIPC group underwent RIPC on the left upper limb twice daily, starting three days before the DCB implantation. The patients were followed up for one year after the operation, and 197 patients returned for coronary angiography (CAG) examination where the quantitative flow ratio (QFR) of the target vessels was measured. The primary endpoint of the study was the incidence of target lesion failure (TLF), which included target lesion revascularization (TLR), target vessel myocardial infarction, and cardiac death. The secondary endpoint was the rate of QFR loss in the target vessels. Results The findings revealed a significantly lower incidence of TLR in the RIPC group compared to the control group. Additionally, at the one-year follow-up, the rate of QFR loss in target vessels was lower in the RIPC group than in the control group. Conclusions The preoperative application of RIPC effectively extended the duration patients could tolerate DCB inflation. Furthermore, this approach positively impacted the long-term prognosis of CAD patients undergoing DCB treatment. Clinical Trial Registration Information NCT04766749.
Collapse
Affiliation(s)
- Zhenzhou Zhao
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan Provincial People’s Hospital, 450003 Zhengzhou, Henan, China
| | - Haosen Yu
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan Provincial People’s Hospital, 450003 Zhengzhou, Henan, China
| | - Ming Nie
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan Provincial People’s Hospital, 450003 Zhengzhou, Henan, China
| | - Xuejie Li
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan Provincial People’s Hospital, 450003 Zhengzhou, Henan, China
| | - Muwei Li
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Henan Provincial People’s Hospital, 450003 Zhengzhou, Henan, China
| |
Collapse
|
3
|
Zhang X, Li B, Yan Y, Sun F, Zhang S, Wang M, Liu H. AT1R autoantibody promotes phenotypic transition of smooth muscle cells by activating AT1R-OAS2. Biochem Pharmacol 2024; 219:115977. [PMID: 38092283 DOI: 10.1016/j.bcp.2023.115977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023]
Abstract
Phenotypic transition of vascular smooth muscle cells (VSMCs) is an early event in the onset and progression of several cardiovascular diseases. As an important mediator of the renin-angiotensin system (RAS), activation of the angiotensin II type 1 receptor (AT1R) induces phenotypic transition of VSMCs. AT1R autoantibodies (AT1-AAs), which are agonistic autoantibodies of AT1R, have been detected in the sera of patients with a variety of cardiovascular diseases associated with phenotypic transition. However, the effect of AT1-AA on phenotypic transition is currently unknown. In this study, AT1-AA-positive rat model was established by active immunization to detect markers of VSMCs phenotypic transition. The results showed that AT1-AA-positive rats showed phenotypic transition of VSMCs, which was evidenced by the decrease of contractile markers, while the increase of synthetic markers in the thoracic aorta. However, in AT1-AA-positive AT1R knockout rats, the phenotypic transition-related proteins were not altered. In vitro, after stimulating human aortic smooth muscle cells with AT1-AA for 48 h, 2'-5' oligoadenylate synthase 2 (OAS2) was identified as the key differentially expressed gene by RNA sequencing and bioinformatics analysis. Furthermore, high expression of OAS2 was found in aorta of AT1-AA-positive rats; knockdown of OAS2 by siRNA can reverse the phenotypic transition of VSMCs induced by AT1-AA. In summary, this study suggests that AT1-AA can promote phenotypic transition of VSMCs through AT1R-OAS2 pathway, and OAS2 might serve as a potential therapeutic target to prevent pathological phenotypic transition of smooth muscle cells.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Bingjie Li
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Yingshuo Yan
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Fei Sun
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China
| | - Meili Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
4
|
Molinuevo MS, Cortizo AM, Sedlinsky C. Effects of advanced glycation end-products, diabetes and metformin on the osteoblastic transdifferentiation capacity of vascular smooth muscle cells: In vivo and in vitro studies. J Diabetes Complications 2023; 37:108626. [PMID: 37839167 DOI: 10.1016/j.jdiacomp.2023.108626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023]
Abstract
AIMS Our objective was to study the vascular smooth muscle cells (VSMC) osteoblastic transdifferentiation in AGE exposed cells or those from diabetic animals, and its response to metformin treatment. METHODS VSMC were obtained from non-diabetic rats, grown with or without AGE; while VSMC of in vivo-ex vivo studies were obtained from non-diabetic control animals (C), diabetic (D), C treated with metformin (M) and D treated with metformin (D-M). We studied the osteoblastic differentiation by evaluating alkaline phosphatase (ALP), type I collagen (Col) and mineral deposit. RESULTS In vitro, AGE increased proliferation, migration, and osteoblastic differentiation of VSMC. Metformin cotreatment prevented the AGE induced proliferation and migration. Both AGE and metformin stimulated the expression of ALP and Col. AGE induced mineralization was prevented by metformin. VSMC from D expressed a higher production of Col and ALP. Those from D-M showed an ALP increase vs C and M, and a partial decrease vs D. Cultured in osteogenic medium, ALP, Col and mineralization increased in D vs C, remained unchanged in M, and were prevented in D-M animals. CONCLUSION Both AGE and DM favor VSMC differentiation towards the osteogenic phenotype and this effect can be prevented by metformin.
Collapse
Affiliation(s)
- María Silvina Molinuevo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina
| | - Ana María Cortizo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina.
| | - Claudia Sedlinsky
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina.
| |
Collapse
|
5
|
Li S, Gao Z, Li H, Xu C, Chen B, Zha Q, Yang K, Wang W. Hif-1α/Slit2 Mediates Vascular Smooth Muscle Cell Phenotypic Changes in Restenosis of Bypass Grafts. J Cardiovasc Transl Res 2023; 16:1021-1031. [PMID: 37097589 PMCID: PMC10615989 DOI: 10.1007/s12265-023-10384-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/24/2023] [Indexed: 04/26/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are involved in restenosis of bypass grafts and cause artery graft occlusion. This study aimed to explore the role of Slit2 in phenotypic switching of VSMCs and its effect on restenosis of vascular conduits. An animal model of vascular graft restenosis (VGR) was produced in SD rats and assessed by echocardiography. The expression of Slit2 and Hif-1α was measured in vivo and in vitro. After Slit2 overexpression, the migration and proliferation of VSMCs were detected in vitro, and the restenosis rates and phenotype of VSMCs were tested in vivo. The arteries of the VGR model presented significant stenosis, and Slit2 was decreased in VSMCs of the VGR model. In vitro, Slit2 overexpression inhibited the migration and proliferation of VSMCs, but Slit2 knockdown promoted migration and proliferation. Hypoxia induced Hif-1α but reduced Slit2, and Hif-1α negatively regulated Slit2 expression. Moreover, Slit2 overexpression weakened the rate of VGR and maintained the patency of artery bypass grafts, which suppressed the phenotypic switching of VSMCs. Slit2 inhibited the synthetic phenotype transformation to inhibit the migration and proliferation of VSMCs and delayed the VGR via Hif-1α.
Collapse
Affiliation(s)
- Sen Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Zhiwei Gao
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Haiqing Li
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Chang Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Bing Chen
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Qing Zha
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Ke Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, Zhejiang, 310009, China.
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
6
|
Teng S, Zhu Z, Wu C, He Y, Zhou S. Inflachromene inhibits intimal hyperplasia through the HMGB1/2- regulated TLR4-NF-κB pathway. Int Immunopharmacol 2023; 119:110198. [PMID: 37087872 DOI: 10.1016/j.intimp.2023.110198] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023]
Abstract
The contractile-syntheticphenotypicconversion of vascular smooth muscle cells (VSMCs) plays a key role in atherosclerosis, vascular restenosis, and hypertension. Our previous study explored the correlation between high mobility group box protein (HMGB) 1 and HMGB2 and neointimal hyperplasia after vascular injury. In the present study, we explore whether inflachromene (ICM), a novel inhibitor of the expression of both HMGB1 and HMGB2, modulates phenotypic changes in VSMCs and the mechanisms involved. Mice treated with ICM after carotid artery wire injury showed a decrease in excessive neointimal hyperplasia compared with that in the vehicle groups. In cultured VSMCs, pretreatment with ICM suppressed the angiotensin II (Ang II)-induced phenotypic conversion, proliferation, and migration. We discovered that ICM reduced the Ang II-induced upregulation of the expression of HMGB1 and HMGB2 and inhibited their shuttling between the nucleus and the cytosol. Mechanistically, Ang II-treated VSMCs exhibited higher levels of Toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) phosphorylation, which were attenuated by ICM. In addition, the NF-κB inhibitor Bay-117082 abolished the recombinant HMGB1-mediated VSMC phenotypic conversion, proliferation, and migration. Furthermore, ICM ameliorated the Ang II-induced increases in NAD[P]H oxidase expression, thereby attenuating the Ang II-induced proliferation and migration. These results reveal that ICM pretreatment attenuates Ang II-induced VSMC dedifferentiation, proliferation, and migration may by regulating the TLR4-NF-kB pathway. Thus, ICM is a potential therapy and preventive treatment for vascular proliferative diseases.
Collapse
Affiliation(s)
- Shuai Teng
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaowei Zhu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chenkai Wu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuhu He
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Relationship between Angiotensin II, Vascular Endothelial Growth Factor, and Arteriosclerosis Obliterans. DISEASE MARKERS 2023; 2023:1316821. [PMID: 36865500 PMCID: PMC9974256 DOI: 10.1155/2023/1316821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/11/2022] [Accepted: 11/24/2022] [Indexed: 02/25/2023]
Abstract
Objective To investigate the relationship between angiotensin II (Ang II), vascular endothelial growth factor (VEGF), and arteriosclerosis obliterans (ASO). Methods 60 ASO patients diagnosed and treated from October 2019 to December 2021 were selected for the observation group while 30 healthy physical examiners were for the control group. The general information (gender, age, history of smoking, diabetes, and hypertension) and arterial blood pressure (systolic and diastolic blood pressure) of the two groups were collected, and parameters like disease site and duration, Fontaine stage, and ankle-brachial index (ABI) of ASO patients have been evaluated. Ang II, VEGF, uric acid (UA), low-density lipoprotein (LDL), high-density lipoprotein (HDL), triglyceride (TG), and total cholesterol (TC) were also detected for the two groups. The variations in UA, LDL, HDL, TG, and TC among two groups along with levels of Ang II and VEGF in ASO patients in accordance to conditions like the general situation, disease duration, disease site, Fontaine stage, and ABI risk level have been studied to establish a correlation between Ang II and VEGF and ASO. Results (1) The proportion of males with a history of smoking, diabetes, and hypertension was higher (P < 0.05) among ASO patients in comparison to the control group. The diastolic blood pressure, LDL, TC, Ang II, and VEGF levels were found to be higher (P < 0.05) whereas HDL was low (P < 0.01). (2) The level of Ang II in male patients with ASO was significantly higher than that in female ASO patients (P < 0.05). In ASO patients, the levels of Ang II and VEGF increased not only with age (P < 0.01) but also with progression in Fontaine stages II, III, and IV (P < 0.01). (3) Logistic regression analysis revealed Ang II and VEGF as risk factors for ASO. (4) An AUC (area under the ROC (receiver operator characteristic) curve) for Ang II and VEGF for the diagnosis of ASO was 0.764 (good) and 0.854 (very good), respectively, while their combined AUC in diagnosing ASO was 0.901 (excellent). The AUC of Ang II and VEGF together in diagnosing ASO was greater than that of Ang II and VEGF alone along with higher specificity as well (all P < 0.05). Conclusion Ang II and VEGF were correlated with the occurrence and development of ASO. The AUC analysis demonstrates that Ang II and VEGF were highly discriminative of ASO.
Collapse
|
8
|
Lu BH, Liu HB, Guo SX, Zhang J, Li DX, Chen ZG, Lin F, Zhao GA. Long non-coding RNAs: Modulators of phenotypic transformation in vascular smooth muscle cells. Front Cardiovasc Med 2022; 9:959955. [PMID: 36093159 PMCID: PMC9458932 DOI: 10.3389/fcvm.2022.959955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022] Open
Abstract
Long non-coding RNA (lncRNAs) are longer than 200 nucleotides and cannot encode proteins but can regulate the expression of genes through epigenetic, transcriptional, and post-transcriptional modifications. The pathophysiology of smooth muscle cells can lead to many vascular diseases, and studies have shown that lncRNAs can regulate the phenotypic conversion of smooth muscle cells so that smooth muscle cells proliferate, migrate, and undergo apoptosis, thereby affecting the development and prognosis of vascular diseases. This review discusses the molecular mechanisms of lncRNA as a signal, bait, stent, guide, and other functions to regulate the phenotypic conversion of vascular smooth muscle cells, and summarizes the role of lncRNAs in regulating vascular smooth muscle cells in atherosclerosis, hypertension, aortic dissection, vascular restenosis, and aneurysms, providing new ideas for the diagnosis and treatment of vascular diseases.
Collapse
Affiliation(s)
- Bing-Han Lu
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Hui-Bing Liu
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
- Henan Normal University, Xinxiang, China
| | - Shu-Xun Guo
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Jie Zhang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Dong-Xu Li
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Zhi-Gang Chen
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Fei Lin
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Guo-An Zhao
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| |
Collapse
|
9
|
Liu JT, Yao QP, Chen Y, Lv F, Liu Z, Bao H, Han Y, Zhang ML, Jiang ZL, Qi YX. Arterial cyclic stretch regulates Lamtor1 and promotes neointimal hyperplasia via circSlc8a1/miR-20a-5p axis in vein grafts. Am J Cancer Res 2022; 12:4851-4865. [PMID: 35836818 PMCID: PMC9274756 DOI: 10.7150/thno.69551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/03/2022] [Indexed: 01/12/2023] Open
Abstract
Rationale: Neointimal hyperplasia caused by dedifferentiation and proliferation of venous smooth muscle cells (SMCs) is the major challenge for restenosis after coronary artery bypass graft. Herein, we investigated the role of Lamtor1 in neointimal formation and the regulatory mechanism of non-coding RNA underlying this process. Methods: Using a "cuff" model, veins were grafted into arterial system and Lamtor1 expression which was correlated with the activation of mTORC1 signaling and dedifferentiation of SMCs, were measured by Western blot. Whole transcriptome deep sequencing (RNA-seq) of the grafted veins combined with bioinformatic analysis identified highly conserved circSlc8a1 and its interaction with miR-20a-5p, which may target Lamtor1. CircSlc8a1 was biochemically characterized by Sanger sequencing and resistant to RNase R digestion. The cytoplasmic location of circSlc8a1 was shown by fluorescence in situ hybridization (FISH). RNA pull-down, luciferase assays and RNA immunoprecipitation (RIP) with Ago2 assays were used to identify the interaction circSlc8a1 with miR-20a-5p. Furthermore, arterial mechanical stretch (10% elongation) was applied in vitro. Results:In vivo, Lamtor1 was significantly enhanced in grafted vein and activated mTORC1 signaling to promote dedifferentiation of SMCs. Arterial mechanical stretch (10% elongation) induced circSlc8a1 expression and positively regulated Lamtor1, activated mTORC1 and promoted SMC dedifferentiation and proliferation. Local injection of circSlc8a1 siRNA or SMC-specific Lamtor1 knockout mice prevented neointimal hyperplasia in vein grafts in vivo. Conclusions: Our study reveals a novel mechanobiological mechanism underlying the dedifferentiation and proliferation of venous SMCs in neointimal hyperplasia. CircSlc81/miR-20a-5p/Lamtor1 axis induced by arterial cyclic stretch may be a potential clinical target that attenuates neointimal hyperplasia in grafted vessels.
Collapse
Affiliation(s)
- Ji-Ting Liu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qing-Ping Yao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Chen
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Lv
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ze Liu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Han Bao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Han
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Liang Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China.,✉ Corresponding authors: Dr. Ying-Xin Qi, Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China. Tel.: +86-21-34204863, Fax: +86-21-34204118, E-mail: ; Dr. Zong-Lai Jiang, Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China. Tel.: +86-21-34204863, Fax: +86-21-34204118, E-mail: ; Dr. Ming-Liang Zhang, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, 600 Yishan Road, Xuhui, Shanghai 200233 China. Tel.: +86-21-24058337, Fax: +86-21-24058337, E-mail:
| | - Zong-Lai Jiang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,✉ Corresponding authors: Dr. Ying-Xin Qi, Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China. Tel.: +86-21-34204863, Fax: +86-21-34204118, E-mail: ; Dr. Zong-Lai Jiang, Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China. Tel.: +86-21-34204863, Fax: +86-21-34204118, E-mail: ; Dr. Ming-Liang Zhang, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, 600 Yishan Road, Xuhui, Shanghai 200233 China. Tel.: +86-21-24058337, Fax: +86-21-24058337, E-mail:
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,✉ Corresponding authors: Dr. Ying-Xin Qi, Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China. Tel.: +86-21-34204863, Fax: +86-21-34204118, E-mail: ; Dr. Zong-Lai Jiang, Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China. Tel.: +86-21-34204863, Fax: +86-21-34204118, E-mail: ; Dr. Ming-Liang Zhang, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, 600 Yishan Road, Xuhui, Shanghai 200233 China. Tel.: +86-21-24058337, Fax: +86-21-24058337, E-mail:
| |
Collapse
|
10
|
Li KX, Wang ZC, Machuki JO, Li MZ, Wu YJ, Niu MK, Yu KY, Lu QB, Sun HJ. Benefits of Curcumin in the Vasculature: A Therapeutic Candidate for Vascular Remodeling in Arterial Hypertension and Pulmonary Arterial Hypertension? Front Physiol 2022; 13:848867. [PMID: 35530510 PMCID: PMC9075737 DOI: 10.3389/fphys.2022.848867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/03/2022] [Indexed: 01/14/2023] Open
Abstract
Growing evidence suggests that hypertension is one of the leading causes of cardiovascular morbidity and mortality since uncontrolled high blood pressure increases the risk of myocardial infarction, aortic dissection, hemorrhagic stroke, and chronic kidney disease. Impaired vascular homeostasis plays a critical role in the development of hypertension-induced vascular remodeling. Abnormal behaviors of vascular cells are not only a pathological hallmark of hypertensive vascular remodeling, but also an important pathological basis for maintaining reduced vascular compliance in hypertension. Targeting vascular remodeling represents a novel therapeutic approach in hypertension and its cardiovascular complications. Phytochemicals are emerging as candidates with therapeutic effects on numerous pathologies, including hypertension. An increasing number of studies have found that curcumin, a polyphenolic compound derived from dietary spice turmeric, holds a broad spectrum of pharmacological actions, such as antiplatelet, anticancer, anti-inflammatory, antioxidant, and antiangiogenic effects. Curcumin has been shown to prevent or treat vascular remodeling in hypertensive rodents by modulating various signaling pathways. In the present review, we attempt to focus on the current findings and molecular mechanisms of curcumin in the treatment of hypertensive vascular remodeling. In particular, adverse and inconsistent effects of curcumin, as well as some favorable pharmacokinetics or pharmacodynamics profiles in arterial hypertension will be discussed. Moreover, the recent progress in the preparation of nano-curcumins and their therapeutic potential in hypertension will be briefly recapped. The future research directions and challenges of curcumin in hypertension-related vascular remodeling are also proposed. It is foreseeable that curcumin is likely to be a therapeutic agent for hypertension and vascular remodeling going forwards.
Collapse
Affiliation(s)
- Ke-Xue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Zi-Chao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | | | - Meng-Zhen Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu-Jie Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ming-Kai Niu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kang-Ying Yu
- Nursing School of Wuxi Taihu University, Wuxi, China
| | - Qing-Bo Lu
- School of Medicine, Southeast University, Nanjing, China
| | - Hai-Jian Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Yes-associated protein reacts differently in vascular smooth muscle cells under different intensities of mechanical stretch. Aging (Albany NY) 2022; 14:286-296. [PMID: 34983026 PMCID: PMC8791225 DOI: 10.18632/aging.203768] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 10/18/2021] [Indexed: 11/25/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are stromal cells of the vascular wall and are continually exposed to mechanical signals. The loss of VSMCs is closely related to the occurrence of many vascular diseases, such as aortic aneurysms and aortic dissection. The proliferation and apoptosis of VSMCs are mechanically stimulated. Yes-associated protein (YAP), one of the core components of the Hippo pathway, plays a key role in the response of VSMCs to mechanical signals. In this study, we tested the impact of different intensities of mechanical stretch on the proliferation and apoptosis of VSMCs, as well as YAP. We tested VSMCs’ proliferation and apoptosis and YAP reaction via immunocytochemistry, western blotting, CCK-8 and flow cytometric analysis. We found that 10% elongation could increase the phosphorylation of YAP and prevent it from entering the nucleus, as well as inhibit cell proliferation and promote apoptosis. However, 15% elongation reduced YAP phosphorylation and promoted its nuclear entry, thereby promoting cell proliferation and inhibiting apoptosis. Accordingly, YAP knockdown suppressed the phenotype of VMSCs induced by 15% elongation. Taken together, YAP regulates proliferation and apoptosis of VSMCs differently under different intensity of mechanical stretch. Mechanical stretch with appropriate intensity can promote the proliferation and inhibit apoptosis of VSMCs by activating YAP.
Collapse
|
12
|
Xie Y, Tan J, Qin Y, Cao Y, Wang Y, Li A, Wang Z, Qiao Z, Yan Z. MiR-3571 modulates the proliferation and migration of vascular smooth muscle cells by targeting claudin 1. Int J Med Sci 2022; 19:511-524. [PMID: 35370461 PMCID: PMC8964332 DOI: 10.7150/ijms.64639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 02/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background and aims: The miRNA-based post-transcription modification has been extensively studied in hypertension. It however remains elusive how miRNA expression is regulated in this pathological process. We hypothesize that hydroxymethylation in the promoter regions tightly controls the levels of key miRNAs, which in turn affects the development of hypertension. Methods: The levels of hydroxymethylation in the promoter regions from thoracic aortic tissues were compared between spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto rats (WKYs), using hydroxymethylcytosine DNA immunoprecipitation (hMeDIP) sequencing. The altered hydroxymethylation level of miR-3571 was confirmed by glucosylation-coupled hydroxymethylation-sensitive qPCR. We further identified claudin 1(CLDN1) as a key target of miR-3571 via bioinformatic prediction (targetscan) and dual-luciferase activity assays. Finally, we analyzed the contribution of miR-3571/CLDN1 axis in the proliferation and migration of vascular smooth muscle cells (VSMCs). Results: The hydroxymethylation level of miR-3571 promoter region in thoracic aortic tissue from spontaneously hypertensive rats was lower than that from normotensive Wistar-Kyoto rats. Accordingly, the expression of miR-3571 was lower during hypertension, with up-regulated CLDN1 protein levels. More importantly, we found that miR3571 overexpression led to phenotypic changes of VSMCs, and inhibited the proliferation and migration of muscle cells via suppressing CLDN1 as well. Our findings further suggested that CLDN1 up-regulation increase the activity of ERK1/2 in VSMCs. Conclusions: Our study suggested that hydroxymethylation in the promoter regions controlled the level of miR-3571 and revealed the important roles of miR-3571 and CLDN1 in VSMCs during the development of hypertension. In addition, our results also indicated that miR-3571/CLDN1 axis regulated the functions of VSMCs via the ERK1/2 pathway. Taken together, our findings support miR-3571 as a novel biomarker for the diagnosis and prevention of hypertension.
Collapse
Affiliation(s)
- Yilin Xie
- Shanghai Jiao Tong University - Minhang Campus, School of Life Science and Biotechnology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai, China
| | - Juanjuan Tan
- Shanghai Jiao Tong University - Minhang Campus, School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composite Materials and Shanghai Key Lab of Electrical Insulation and Thermal Ageing, Shanghai, China
| | - Yingchun Qin
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong Cao
- Shanghai Jiao Tong University School of Life Sciences and Biotechnology, Shanghai, China
| | - Yicheng Wang
- Shanghai Jiao Tong University School of Life Sciences and Biotechnology, Shanghai, China
| | - Aihua Li
- Anhui University of Science and Technology, Huainan, Anhui, China
| | - Zhaoxia Wang
- Shanghai Jiao Tong University Laboratory Animal Center, Shanghai, China
| | - Zhongdong Qiao
- Shanghai Jiao Tong University School of Life Sciences and Biotechnology, Shanghai, China
| | - Zhiqiang Yan
- Shanghai Jiao Tong University affiliated sixth people's hospital south campus, Central Laboratory, Shanghai, China
| |
Collapse
|
13
|
PDE-Mediated Cyclic Nucleotide Compartmentation in Vascular Smooth Muscle Cells: From Basic to a Clinical Perspective. J Cardiovasc Dev Dis 2021; 9:jcdd9010004. [PMID: 35050214 PMCID: PMC8777754 DOI: 10.3390/jcdd9010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases are important causes of mortality and morbidity worldwide. Vascular smooth muscle cells (SMCs) are major components of blood vessels and are involved in physiologic and pathophysiologic conditions. In healthy vessels, vascular SMCs contribute to vasotone and regulate blood flow by cyclic nucleotide intracellular pathways. However, vascular SMCs lose their contractile phenotype under pathological conditions and alter contractility or signalling mechanisms, including cyclic nucleotide compartmentation. In the present review, we focus on compartmentalized signaling of cyclic nucleotides in vascular smooth muscle. A deeper understanding of these mechanisms clarifies the most relevant axes for the regulation of vascular tone. Furthermore, this allows the detection of possible changes associated with pathological processes, which may be of help for the discovery of novel drugs.
Collapse
|
14
|
He Y, Wang R, Zhang P, Yan J, Gong N, Li Y, Dong S. Curcumin inhibits the proliferation and migration of vascular smooth muscle cells by targeting the chemerin / CMKLR1 / LCN2 axis. Aging (Albany NY) 2021; 13:13859-13875. [PMID: 34029211 PMCID: PMC8202847 DOI: 10.18632/aging.202980] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 01/20/2021] [Indexed: 12/31/2022]
Abstract
Atherosclerosis (AS) is a chronic progressive inflammatory disease and a leading cause of death worldwide. Being a novel adipokine, chemerin is reported to be positively correlated with the severity of AS, yet its underlying mechanisms in AS remains elusive. It is well-known that AS development is significantly attributed to abnormal proliferation and migration of vascular smooth muscle cells (VSMCs). Therefore, we investigated the role of the chemerin / chemokine-like receptor 1 (CMKLR1, chemerin receptor) signaling, and the potential therapeutic effect of curcumin in VSMCs proliferation and migration during AS by establishing a high fat diet (HFD) mouse model. We found that CMKLR1 was highly expressed in HFD-induced AS tissues and that its expression level was positively correlated with aortic proliferation. Knockdown of CMKLR1 significantly inhibited VSMCs proliferation and migration, as evidenced by the EdU-incorporation assay, wound healing assay, and the induction of proliferating cell nuclear antigen (PCNA) and matrix metalloproteinase-9 (MMP-9) expression. Furthermore, we discovered that Lipocalin-2 (LCN2) acts as a key factor involved in CMKLR1-mediated VSMCs proliferation and migration via the p38 / MAPK and Wnt / β-catenin signaling pathways, and we demonstrated that curcumin inhibits VSMCs proliferation and migration by inhibiting chemerin / CMKLR1 / LCN2, thereby reducing AS progression. Our findings suggest that chemerin / CMKLR1 activation promotes the development of AS; hence, targeting the chemerin / CMKLR1 / LCN2 signaling pathway may be a reasonable treatment modality for AS.
Collapse
Affiliation(s)
- Yaqiong He
- Department of Cardiology, Shenzhen People’s Hospital, Jinan University, Shenzhen 518000, Guangdong, China
| | - Rongning Wang
- Department of Cardiology, Shenzhen People’s Hospital, Jinan University, Shenzhen 518000, Guangdong, China
| | - Peng Zhang
- Department of Cardiology, Shenzhen People’s Hospital, Jinan University, Shenzhen 518000, Guangdong, China
| | - Jianlong Yan
- Department of Cardiology, Shenzhen People’s Hospital, Jinan University, Shenzhen 518000, Guangdong, China
| | - Nan Gong
- Department of Cardiology, Shenzhen People’s Hospital, Jinan University, Shenzhen 518000, Guangdong, China
| | - Yuhang Li
- Department of Orthopedics, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong, China
| | - Shaohong Dong
- Department of Cardiology, Shenzhen People’s Hospital, Jinan University, Shenzhen 518000, Guangdong, China
| |
Collapse
|
15
|
Qi HM, Cao Q, Liu Q. TLR4 regulates vascular smooth muscle cell proliferation in hypertension via modulation of the NLRP3 inflammasome. Am J Transl Res 2021; 13:314-325. [PMID: 33527026 PMCID: PMC7847527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/11/2020] [Indexed: 06/12/2023]
Abstract
UNLABELLED Backgroud: Toll-like receptor 4 (TLR4), a key mediator of inflammatory responses, which is associated with vascular remodeling. The association between TLR4 and NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome in the regulation of vascular smooth muscle cell (VSMC) proliferation remains unclear. This study was to explore the role and underlying mechanisms of TLR4 in the proliferation of VSMC in hypertension. METHODS VSMC proliferation after TLR4 overexpression or downregulation was determined by CCK-8, EdU Incorporation and colony formation assays. Western blots were carried out to investigate the expression of TLR4 and NLRP3 inflammasome components in VSMCs. Next, blood pressure measurements and Hematoxylin and Eosin (HE) staining assays were performed in spontaneously hypertensive rats (SHR). Media thickness (M) and diameter lumen (L) were measured as indicators of vascular remodeling. The expression of TLR4, PCNA and NLRP3 inflammasome complex was analyzed by Western blots in the aorta of SHR. RESULTS We showed that TLR4 overexpression with cDNA enhanced, while knockdown of TLR4 with shRNA inhibited Ang II-induced VSMC proliferation. Besides, TLR4 overexpression upregulated the proteion expression of the NLRP3 inflammasome components including NLRP3, ASC and caspase-1, whereas their corresponding levels of expression were observed to decrease in TLR4 shRNA-transfected VSMCs. Knockdown of TLR4 attenuated vascular remodeling, blood pressure (BP) and the levels of NLRP3, ASC, caspase-1, IL-1β and IL-18 in SHR aortas. CONCLUSION This study revealed that TLR4 regulated Ang II-induced VSMC proliferation through modulating the NLRP3 inflammasome. Knockdown of TLR4 attenuated the BP and vascular remodeling by inhibiting the expression of the NLRP3 inflammasome component in SHR. Our results support that TLR4 regulates VSMC proliferation in hypertension via triggering the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Hui-Meng Qi
- Department of General Practice, The First Hospital of China Medical UniversityShenyang, China
| | - Qin Cao
- Department of Gastroenterolog, The First Hospital of China Medical UniversityShenyang, China
| | - Qiang Liu
- Department of Nephrology, The First Hospital of China Medical UniversityShenyang, China
| |
Collapse
|
16
|
Wang Y, Huang XX, Leng D, Li JF, Liang Y, Jiang T. Effect of EZH2 on pulmonary artery smooth muscle cell migration in pulmonary hypertension. Mol Med Rep 2020; 23:129. [PMID: 33313943 PMCID: PMC7751464 DOI: 10.3892/mmr.2020.11768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 10/27/2020] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a life‑threatening disease that often involves vascular remodeling. Although pulmonary arterial smooth muscle cells (PASMCs) are the primary participants in vascular remodeling, their biological role is not entirely clear. The present study analyzed the role of enhancer of zeste homolog 2 (EZH2) in vascular remodeling of PH by investigating the behavior of PASMCs. The expression levels of EZH2 in PASMCs in chronic thromboembolic pulmonary hypertension (CTEPH), a type of PH, were detected. The role of EZH2 in PASMC migration was investigated by wound‑healing assay following overexpression and knockdown. Functional enrichment analysis of the whole‑genome expression profiles of PASMCs with EZH2 overexpression was performed using an mRNA Human Gene Expression Microarray. Quantitative (q)PCR was performed to confirm the results of the microarray. EZH2 expression levels increased in CTEPH cell models. The overexpression of EZH2 enhanced PASMC migration compared with control conditions. Functional enrichment analysis of the differentially expressed genes following EZH2 overexpression indicated a strong link between EZH2 and the immune inflammatory response and oxidoreductase activity in PASMCs. mRNA expression levels of superoxide dismutase 3 were verified by qPCR. The results suggested that EZH2 was involved in the migration of PASMCs in PH, and may serve as a potential target for the treatment of PH.
Collapse
Affiliation(s)
- Ying Wang
- Department of Clinical Laboratory, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiao-Xi Huang
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine, Beijing 100020, P.R. China
| | - Dong Leng
- Department of Clinical Laboratory, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Ji-Feng Li
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine, Beijing 100020, P.R. China
| | - Yan Liang
- Department of Clinical Laboratory, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
17
|
LncRNA BANCR induced vascular smooth muscle cell proliferation by downregulating miR-34c methylation in atherosclerosis. J Thromb Thrombolysis 2020; 51:924-932. [PMID: 33151462 DOI: 10.1007/s11239-020-02314-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 02/07/2023]
Abstract
Aberrant vascular smooth muscle cell (VSMCs) proliferation involves in the development of atherosclerosis. It reported that Long noncoding BRAF-activated noncoding RNA (BANCR) and miR-34c played opposite roles in the regulation of the proliferation of VSMCs, indicating that there might be a potential interaction between them. This study was to investigate the relationship between BANCR and miR-34c in atherosclerosis. Blood (5 ml) was obtained from 56 patients with atherosclerosis and 56 healthy volunteers after they were fasted overnight, and plasma was extracted from the blood. Human Aortic Smooth Muscle Cells (HASMCs) were used to perform in vitro cell experiments. RT-qPCR was performed to measure the expression of BANCR and miR-34c in plasma and HASMCs. Dual luciferase reporter assay detected the interaction between BANCR and miR-34c. CCK-8 assay was used to assess the effects of BANCR and miR-34c overexpression on the proliferation of HASMCs. Western blotting was used to assess the effects of BANCR and miR-34c overexpression on the protein expression of HMGB1, TNF-ɑ and Bcl-2. In this study, we found that BANCR was upregulated, while miR-34c was downregulated in atherosclerosis. Bioinformatics analysis showed that BANCR and miR-34c could directly interact with each other. Moreover, overexpression of BANCR could decrease the expression of miR-34c in HASMCs, but overexpression of miR-34c could not affect the expression of BANCR. Furthermore, overexpression of BACNR increased miR-34c methylation, and knockdown of endogenous BANCR decreased miR-34c methylation. In addition, overexpression of BANCR reduced the effects of miR-34c on HASMCs proliferation and reversed the effects of miR-34c on HMGB1, TNF-ɑ and Bcl-2 expression. BANCR overexpression could induce HASMCs proliferation by downregulating the miR-34c methylation. Therefore given BANCR upregulation in atherosclerosis, its expression may be considered as a novel and useful biomarker for atherosclerosis prevention and prognosis. However considering the possible effects of other underlying diseases on both BANCR expression and miR-34c in atherosclerosis, further investigation is suggested for future research.
Collapse
|
18
|
Seong JH, Song YS, Joo HW, Park IH, Shen GY, Shin NK, Lee AH, Kwon AM, Lee Y, Kim H, Kim KS. Modified method for effective primary vascular smooth muscle progenitor cell culture from peripheral blood. Cytotechnology 2020; 72:763-772. [PMID: 32909140 PMCID: PMC7547929 DOI: 10.1007/s10616-020-00419-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 09/02/2020] [Indexed: 11/24/2022] Open
Abstract
In previous studies, vascular smooth muscle progenitor cells (vSMPCs) isolated from peripheral blood mononuclear cells (PBMCs) were cultured using medium containing platelet-derived growth factor-BB (PDGF-BB) for 4 weeks. However, this method requires long culture periods of up to 4 weeks and yields low cell counts. Therefore, we proposed the modified method to improve the cell yield and purity and to reduce the cell culture period. PBMCs were isolated from human peripheral blood and cultured by the conventional method using medium containing PDGF-BB alone or the modified method using medium containing PDGF-BB, basic fibroblast growth factor (bFGF), and insulin-transferrin-selenium ITS for 4 weeks. The purity of vSMPCs was analyzed for the expression of a- smooth muscle actin (SMA) by flow cytometry and significantly higher in the modified method than conventional methods at the 1st and 2nd weeks. Also, mRNA expression of a-SMA by real-time PCR was significantly higher in the modified method than conventional method at the 2 weeks. The yield of vSMPCs by trypan blue exclusion assay was significantly higher in the modified method than conventional method at the 1st, 2nd and 3rd weeks. The primary culture using the modified method with PDGF-BB, bFGF, and ITS not only improved cell purity and yield, but also shortened the culture period, compared to the conventional culture method for vSMPCs. The modified method will be a time-saving and useful tool in various studies related to vascular pathology.
Collapse
Affiliation(s)
- Jin-Hee Seong
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Yi-Sun Song
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Hyun-Woo Joo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - In-Hwa Park
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Guang-Yin Shen
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
- Division of Cardiology, Department of Internal Medicine, Jilin University Jilin Central Hospital, Jilin, China
| | - Na-Kyoung Shin
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - A-Hyeon Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Amy M Kwon
- Biostatistical Consulting and Research Laboratory, Medical Research Collaborating Center, Industry-University Cooperation Foundation, Hanyang University, Seoul, South Korea
| | - Yonggu Lee
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea
| | - Hyuck Kim
- Department of Thoracic Surgery, Hanyang University Seoul Hospital, Seoul, South Korea
| | - Kyung-Soo Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea.
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea.
| |
Collapse
|
19
|
He X, Lian Z, Yang Y, Wang Z, Fu X, Liu Y, Li M, Tian J, Yu T, Xin H. Long Non-coding RNA PEBP1P2 Suppresses Proliferative VSMCs Phenotypic Switching and Proliferation in Atherosclerosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:84-98. [PMID: 32916601 PMCID: PMC7490454 DOI: 10.1016/j.omtn.2020.08.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/31/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022]
Abstract
Long non-coding RNAs (lncRNAs) play a crucial role in the growth of vascular smooth muscle cells (VSMCs), the dysfunction of which is closely associated with the initiation and progression of cardiovascular diseases (CVDs). Abnormal phenotypic switching and proliferation of VSMCs constitute a significant event in the progression of atherosclerosis. The present study identified a novel lncRNA, PEBP1P2, which serves as a valuable regulator of VSMCs in phenotypic transformation and proliferation. The expression of PEBP1P2 was remarkably decreased in proliferating VSMCs and pathological arteries when using a balloon injury model of rats. Furthermore, we found that PEBP1P2 represses proliferation, migration, and dedifferentiation during phenotype switching in VSMCs induced by platelet-derived growth factor BB (PDGF-BB). Mechanistically, cyclin-dependent kinase 9 (CDK9) was confirmed to be the direct target of PEBP1P2, which was proven to mediate phenotypic switching and proliferation of VSMCs and was rescued by PEBP1P2. Then, we explored the clinical significance, as we observed the decreased expression of PEBP1P2 in the serum of coronary heart disease (CHD) patients and human advanced carotid atherosclerotic plaques. Finally, PEBP1P2 overexpression distinctly suppressed neointima formation and VSMC phenotypic switching in vivo. Taken together, PEBP1P2 inhibits proliferation and migration in VSMCs by directly binding to CDK9, implying that it may be a promising therapeutic target for the treatment of proliferative vascular diseases.
Collapse
Affiliation(s)
- Xingqiang He
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China; Department of Cardiology, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi, P.R. China
| | - Zhexun Lian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, P.R. China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China
| | - Yan Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266021, Shandong, P.R. China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266021, Shandong, P.R. China
| | - Jiawei Tian
- Department of Emergency Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266021, Shandong, P.R. China; Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China.
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, P.R. China.
| |
Collapse
|
20
|
Hughes WE, Beyer AM, Gutterman DD. Vascular autophagy in health and disease. Basic Res Cardiol 2020; 115:41. [PMID: 32506214 DOI: 10.1007/s00395-020-0802-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Homeostasis is maintained within organisms through the physiological recycling process of autophagy, a catabolic process that is intricately involved in the mobilization of nutrients during starvation, recycling of cellular cargo, as well as initiation of cellular death pathways. Specific to the cardiovascular system, autophagy responds to both chemical (e.g. free radicals) and mechanical stressors (e.g. shear stress). It is imperative to note that autophagy is not a static process, and measurement of autophagic flux provides a more comprehensive investigation into the role of autophagy. The overarching themes emerging from decades of autophagy research are that basal levels of autophagic flux are critical, physiological stressors may increase or decrease autophagic flux, and more importantly, aberrant deviations from basal autophagy may elicit detrimental effects. Autophagy has predominantly been examined within cardiac or vascular smooth muscle tissue within the context of disease development and progression. Autophagic flux within the endothelium holds an important role in maintaining vascular function, demonstrated by the necessary role for intact autophagic flux for shear-induced release of nitric oxide however the underlying mechanisms have yet to be elucidated. Within this review, we theorize that autophagy itself does not solely control vascular homeostasis, rather, it works in concert with mitochondria, telomerase, and lipids to maintain physiological function. The primary emphasis of this review is on the role of autophagy within the human vasculature, and the integrative effects with physiological processes and diseases as they relate to the vascular structure and function.
Collapse
Affiliation(s)
- William E Hughes
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA.
| | - Andreas M Beyer
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA
| | - David D Gutterman
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA
| |
Collapse
|
21
|
Balčiūnas E, Dreižė N, Grubliauskaitė M, Urnikytė S, Šimoliūnas E, Bukelskienė V, Valius M, Baldock SJ, Hardy JG, Baltriukienė D. Biocompatibility Investigation of Hybrid Organometallic Polymers for Sub-Micron 3D Printing via Laser Two-Photon Polymerisation. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E3932. [PMID: 31783647 PMCID: PMC6926539 DOI: 10.3390/ma12233932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/20/2019] [Accepted: 11/24/2019] [Indexed: 01/20/2023]
Abstract
Hybrid organometallic polymers are a class of functional materials which can be used to produce structures with sub-micron features via laser two-photon polymerisation. Previous studies demonstrated the relative biocompatibility of Al and Zr containing hybrid organometallic polymers in vitro. However, a deeper understanding of their effects on intracellular processes is needed if a tissue engineering strategy based on these materials is to be envisioned. Herein, primary rat myogenic cells were cultured on spin-coated Al and Zr containing polymer surfaces to investigate how each material affects the viability, adhesion strength, adhesion-associated protein expression, rate of cellular metabolism and collagen secretion. We found that the investigated surfaces supported cellular growth to full confluency. A subsequent MTT assay showed that glass and Zr surfaces led to higher rates of metabolism than did the Al surfaces. A viability assay revealed that all surfaces supported comparable levels of cell viability. Cellular adhesion strength assessment showed an insignificantly stronger relative adhesion after 4 h of culture than after 24 h. The largest amount of collagen was secreted by cells grown on the Al-containing surface. In conclusion, the materials were found to be biocompatible in vitro and have potential for bioengineering applications.
Collapse
Affiliation(s)
- Evaldas Balčiūnas
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| | - Nadežda Dreižė
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| | - Monika Grubliauskaitė
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| | - Silvija Urnikytė
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| | - Egidijus Šimoliūnas
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| | - Virginija Bukelskienė
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| | - Mindaugas Valius
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| | - Sara J. Baldock
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, UK;
- Materials Science Institute, Lancaster University, Lancaster LA1 4YB, UK
| | - John G. Hardy
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, UK;
- Materials Science Institute, Lancaster University, Lancaster LA1 4YB, UK
| | - Daiva Baltriukienė
- Institute of Biochemistry, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania; (E.B.); (N.D.); (M.G.); (S.U.); (E.Š.); (V.B.); (M.V.)
| |
Collapse
|
22
|
Wang Y, Zhang X, Gao L, Li J, Chen W, Chi J, Zhang X, Fu Y, Zhao M, Liu N, Li Y, Xu Y, Yang K, Yin X, Liu Y. Cortistatin exerts antiproliferation and antimigration effects in vascular smooth muscle cells stimulated by Ang II through suppressing ERK1/2, p38 MAPK, JNK and ERK5 signaling pathways. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:561. [PMID: 31807542 DOI: 10.21037/atm.2019.09.45] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Vascular remodeling, that contributes to cardiovascular diseases such as hypertension develops by anomalous proliferation and migration of vascular smooth muscle cells (VSMCs). Cortistatin (CST), a newly discovered biological peptide, has been acknowledged for its protective effects against cardiovascular diseases. Whether CST has an inhibitory regulation role in angiotensin II (Ang II)-induced proliferation and migration of VSMCs and what molecular mechanisms may participate in the CST inhibition process are still unknown. Methods VSMCs were divided into control group, Ang II (10-7 M) group, Ang II + PD98059 (5×10-5 M) group, Ang II + SB203580 (10-5 M) group, Ang II + SP600125 (10-5 M) group, Ang II + XMD17-109 (10-6 M) group, Ang II + CST (10-8 M) group and Ang II + CST (10-7 M) group. Cell proliferation was detected by western blotting and cell counting kit-8 (CCK8) analysis. Migration of VSMCs was measured by Transwell assay. Results Compared with control group, Ang II upregulated the expression levels of proliferating cell nuclear antigen (PCNA) and osteopontin (OPN) and downregulated that of α-smooth muscle actin (α-SMA), increased the proliferation rate as shown by CCK8 and VSMC migration as shown by Transwell assay in cultured VSMCs of the Ang II group. Meanwhile, in Ang II-cultured VSMCs, we found activation of extracellular signal-regulated kinase (ERK) 1/2, p38 MAP kinase (p38 MAPK), c-Jun N-terminal kinase (JNK), and ERK5 pathways by western blotting at different time points. However, the proliferation and migration stimulated by Ang II were partly reversed by drug inhibitors of the four pathways, namely, PD98059, SB203580, SP600125 and XMD17-109. When Ang II-stimulated VSMCs were cultured with CST pretreatment, we found that proliferation and migration were greatly suppressed as well as that the ERK1/2, p38 MAPK, JNK and ERK5 pathways were deactivated by CST. Conclusions The accumulated data suggest that CST may play a protective role in Ang II-promoted proliferation and migration of VSMCs via inhibiting the mitogen-activated protein kinase (MAPK) family pathways, providing a new orientation of CST in protecting against cardiovascular diseases.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin Zhang
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lei Gao
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jihe Li
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wenjia Chen
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jinyu Chi
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xiaohui Zhang
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yu Fu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Meng Zhao
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Na Liu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yang Li
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yang Xu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Kelaier Yang
- Department of Endocrine, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xinhua Yin
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yue Liu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
23
|
Prolyl 4-Hydroxylase Domain Protein 3-Inhibited Smooth-Muscle-Cell Dedifferentiation Improves Cardiac Perivascular Fibrosis Induced by Obstructive Sleep Apnea. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9174218. [PMID: 31346526 PMCID: PMC6621170 DOI: 10.1155/2019/9174218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/10/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Background Intermittent hypoxia (IH) induced by obstructive sleep apnea (OSA) is a leading factor affecting cardiovascular fibrosis. Under IH condition, smooth muscle cells (SMAs) respond by dedifferentiation, which is associated with vascular remodelling. The expression of prolyl 4-hydroxylase domain protein 3 (PHD3) increases under hypoxia. However, the role of PHD3 in OSA-induced SMA dedifferentiation and cardiovascular fibrosis remains uncertain. Methods We explored the mechanism of cardiovascular remodelling in C57BL/6 mice exposed to IH for 3 months and investigated the mechanism of PHD3 in improving the remodelling in vivo and vitro. Results In vivo remodelling showed that IH induced cardiovascular fibrosis via SMC dedifferentiation and that fibrosis improved when PHD3 was overexpressed. In vitro remodelling showed that IH induced SMA dedifferentiation, which secretes much collagen I. PHD3 overexpression in cultured SMCs reversed the dedifferentiation by degrading and inactivating HIF-1α. Conclusion OSA-induced cardiovascular fibrosis was associated with SMC dedifferentiation, and PHD3 overexpression may benefit its prevention by reversing the dedifferentiation. Therefore, PHD3 overexpression has therapeutic potential in disease treatment.
Collapse
|
24
|
Wadey K, Lopes J, Bendeck M, George S. Role of smooth muscle cells in coronary artery bypass grafting failure. Cardiovasc Res 2019; 114:601-610. [PMID: 29373656 DOI: 10.1093/cvr/cvy021] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/22/2018] [Indexed: 01/30/2023] Open
Abstract
Atherosclerosis is the underlying pathology of many cardiovascular diseases. The formation and rupture of atherosclerotic plaques in the coronary arteries results in angina and myocardial infarction. Venous coronary artery bypass grafts are designed to reduce the consequences of atherosclerosis in the coronary arteries by diverting blood flow around the atherosclerotic plaques. However, vein grafts suffer a high failure rate due to intimal thickening that occurs as a result of vascular cell injury and activation and can act as 'a soil' for subsequent atherosclerotic plaque formation. A clinically-proven method for the reduction of vein graft intimal thickening and subsequent major adverse clinical events is currently not available. Consequently, a greater understanding of the underlying mechanisms of intimal thickening may be beneficial for the design of future therapies for vein graft failure. Vein grafting induces inflammation and endothelial cell damage and dysfunction, that promotes vascular smooth muscle cell (VSMC) migration, and proliferation. Injury to the wall of the vein as a result of grafting leads to the production of chemoattractants, remodelling of the extracellular matrix and cell-cell contacts; which all contribute to the induction of VSMC migration and proliferation. This review focuses on the role of altered behaviour of VSMCs in the vein graft and some of the factors which critically lead to intimal thickening that pre-disposes the vein graft to further atherosclerosis and re-occurrence of symptoms in the patient.
Collapse
Affiliation(s)
- Kerry Wadey
- Bristol Medical School, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Joshua Lopes
- Translational Biology and Engineering Program, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Michelle Bendeck
- Translational Biology and Engineering Program, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Sarah George
- Bristol Medical School, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| |
Collapse
|
25
|
Li FJ, Zhang CL, Luo XJ, Peng J, Yang TL. Involvement of the MiR-181b-5p/HMGB1 Pathway in Ang II-induced Phenotypic Transformation of Smooth Muscle Cells in Hypertension. Aging Dis 2019; 10:231-248. [PMID: 31011475 PMCID: PMC6457049 DOI: 10.14336/ad.2018.0510] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/10/2018] [Indexed: 12/16/2022] Open
Abstract
Phenotypic transformation of vascular smooth muscle cells (VSMCs) contributes to vascular remodeling in hypertension. High mobility group box-1 (HMGB1) has been reported to be involved in several pathogenic processes including VSMC proliferation and migration. The present study was designed to determine the role of HMGB1 in VSMC phenotypic transformation in hypertension. First, we demonstrated that HMGB1 was elevated in a model of Ang II-induced VSMC phenotypic transformation, which showed down-regulation of contractile proteins and up-regulation of synthetic proteins. Knockdown of HMGB1 and losartan could block the phenotypic transformation. Next, we identified three potential miRNAs for upstream regulation of HMGB1 by bioinformatic analysis; only miR-181b-5p was significantly down-regulated in Ang II-treated cells. Co-treating the cells with miR-181b-5p mimics suppressed HMGB1 expression as well as the phenotypic transformation, migration, and proliferation. Furthermore, the luciferase reporter gene assay confirmed the direct interaction between miR-181b-5p and HMGB1. Finally, to extend these cell-based studies to clinical patients, we demonstrated that plasma miR-181b-5p levels were decreased, while Ang II and HMGB1 levels, as well as the intima-media thickness (IMT) were increased in hypertensive patients; these effects were reversed following the administration of angiotensin receptor blockers. Based on these observations, we conclude that the down-regulation of miR-181b-5p leads to the elevation of HMGB1 levels in hypertensive patients, which accounts, at least partially, for VSMCs phenotypic transformation and vascular remodeling. Our findings also highlight that the plasma levels of miR-181b-5p and HMGB1 may serve as novel biomarkers for vascular remodeling in the hypertensive patients.
Collapse
Affiliation(s)
- Feng-Juan Li
- 1Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Cheng-Long Zhang
- 1Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiu-Ju Luo
- 2Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha410013, China
| | - Jun Peng
- 3Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,4Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Tian-Lun Yang
- 1Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
26
|
Liu LB, Shen HF, Cha W, Jin ZJ, Xia HJ, Liu JJ, Hu JF. SXBX pill suppresses homocysteine-induced vascular smooth muscle cells dedifferentiation by inhibiting NLRP3 inflammasomes activation via ERK/p38 MAPK pathways. Am J Transl Res 2019; 11:806-818. [PMID: 30899381 PMCID: PMC6413280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 12/28/2018] [Indexed: 06/09/2023]
Abstract
The dedifferentiation of vascular smooth muscle cells (VSMCs) is a key event in the pathogenesis of vascular remodeling-related disease. The present study aimed to investigate the effects of shexiangbaoxin (SXBX) pill, a traditional Chinese medicinal formula on VSMCs dedifferentiation and its potential mechanisms. High-fat diet (HFD) was introduced to lipoprotein receptor-deficient (LDLR-/-) mice to generate hyperhomocysteinemia (HHcy), and plasma Hcy and lipid levels were analyzed. The phenotype of VSMCs was assessed in mice with the treatment of low (45 mg/kg/d) or high (90 mg/kg/d) SXBX pill by measuring the contractile protein α-SMA, SM22α and synthetic proteins OPN using RT-qPCR, western blotting and immunofluorescence assay. In vitro, the proliferation, migration and dedifferentiation of VSMCs were measured by MTT, Edu incorporation, wound healing and western blotting assay. Small interfering RNA technology was used to examine the role of NLRP3 in the effects of SXBX pill on dedifferentiation. The results indicated that although SXBX pill had no influence on HFD-induced HHcy and hyperlipidaemia, it reversed HHcy-induced dedifferentiation of VSMCs in vivo. SXBX pill significantly inhibited proliferation, migration and dedifferentiation of Hcy-treated VSMCs. In addition, we found that Hcy activated NLRP3 inflammasomes in VSMCs and SXBX pill could attenuate NLRP3 inflammasomes activation. Moreover, subsequent analysis suggested that SXBX pill inhibited NLRP3 inflammasomes activation through regulation of ERK1/2 and p38 MAPK pathway. Knockdown of NLRP3 reversed the inhibitory effects of SXBX pill in VSMCs. In conclusion, SXBX pill inhibited Hcy-induced proliferation, migration and dedifferentiation of VSMCs by suppressing NLRP3 inflammasomes activation via of ERK/p38 MAPK pathway.
Collapse
Affiliation(s)
- Long Bin Liu
- Department of Cardiology, The Affiliated Hospital of Shaoxing UniversityShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing Municipal HospitalShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang Province, China
| | - Hong Feng Shen
- Department of Cardiology, The Affiliated Hospital of Shaoxing UniversityShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing Municipal HospitalShaoxing, Zhejiang Province, China
| | - Wei Cha
- Department of Cardiology, The Affiliated Hospital of Shaoxing UniversityShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing Municipal HospitalShaoxing, Zhejiang Province, China
| | - Zhi Jiang Jin
- Department of Cardiology, The Affiliated Hospital of Shaoxing UniversityShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing Municipal HospitalShaoxing, Zhejiang Province, China
| | - Hai Jiang Xia
- Department of Cardiology, The Affiliated Hospital of Shaoxing UniversityShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing Municipal HospitalShaoxing, Zhejiang Province, China
| | - Jing Jing Liu
- Department of Cardiology, The Affiliated Hospital of Shaoxing UniversityShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing Municipal HospitalShaoxing, Zhejiang Province, China
| | - Jia Feng Hu
- Department of Cardiology, The Affiliated Hospital of Shaoxing UniversityShaoxing, Zhejiang Province, China
- Department of Cardiology, Shaoxing Municipal HospitalShaoxing, Zhejiang Province, China
| |
Collapse
|
27
|
BCL6 Attenuates Proliferation and Oxidative Stress of Vascular Smooth Muscle Cells in Hypertension. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5018410. [PMID: 30805081 PMCID: PMC6362478 DOI: 10.1155/2019/5018410] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Proliferation and oxidative stress of vascular smooth muscle cells (VSMCs) contribute to vascular remodeling in hypertension and several major vascular diseases. B-cell lymphoma 6 (BCL6) functions as a transcriptional repressor. The present study is designed to determine the roles of BCL6 in VSMC proliferation and oxidative stress and underlying mechanism. Angiotensin (Ang) II was used to induce VSMC proliferation and oxidative stress in human VSMCs. Effects of BCL6 overexpression and knockdown were, respectively, investigated in Ang II-treated human VSMCs. Therapeutical effects of BCL6 overexpression on vascular remodeling, oxidative stress, and proliferation were determined in the aorta of spontaneously hypertensive rats (SHR). Ang II reduced BCL6 expression in human VSMCs. BCL6 overexpression attenuated while BCL6 knockdown enhanced the Ang II-induced upregulation of NADPH oxidase 4 (NOX4), production of reactive oxygen species (ROS), and proliferation of VSMCs. BCL6 expression was downregulated in SHR. BCL6 overexpression in SHR reduced NOX4 expression, ROS production, and proliferation of the aortic media of SHR. Moreover, BCL6 overexpression attenuated vascular remodeling and hypertension in SHR. However, BCL6 overexpression had no significant effects on NOX2 expression in human VSMCs or in SHR. We conclude that BCL6 attenuates proliferation and oxidative stress of VSMCs in hypertension.
Collapse
|
28
|
van Keulen D, Pouwer MG, Pasterkamp G, van Gool AJ, Sollewijn Gelpke MD, Princen HMG, Tempel D. Inflammatory cytokine oncostatin M induces endothelial activation in macro- and microvascular endothelial cells and in APOE*3Leiden.CETP mice. PLoS One 2018; 13:e0204911. [PMID: 30273401 PMCID: PMC6166945 DOI: 10.1371/journal.pone.0204911] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 09/17/2018] [Indexed: 01/08/2023] Open
Abstract
AIMS Endothelial activation is involved in many chronic inflammatory diseases, such as atherosclerosis, and is often initiated by cytokines. Oncostatin M (OSM) is a relatively unknown cytokine that has been suggested to play a role in both endothelial activation and atherosclerosis. We comprehensively investigated the effect of OSM on endothelial cell activation from different vascular beds and in APOE*3Leiden.CETP mice. METHODS AND RESULTS Human umbilical vein endothelial cells, human aortic endothelial cells and human microvascular endothelial cells cultured in the presence of OSM express elevated MCP-1, IL-6 and ICAM-1 mRNA levels. Human umbilical vein endothelial cells and human aortic endothelial cells additionally expressed increased VCAM-1 and E-selectin mRNA levels. Moreover, ICAM-1 membrane expression is increased as well as MCP-1, IL-6 and E-selectin protein release. A marked increase was observed in STAT1 and STAT3 phosphorylation indicating that the JAK/STAT pathway is involved in OSM signaling. OSM signals through the LIF receptor alfa (LIFR) and the OSM receptor (OSMR). siRNA knockdown of the LIFR and the OSMR revealed that simultaneous knockdown is necessary to significantly reduce MCP-1 and IL-6 secretion, VCAM-1 and E-selectin shedding and STAT1 and STAT3 phosphorylation after OSM stimulation. Moreover, OSM administration to APOE*3Leiden.CETP mice enhances plasma E-selectin levels and increases ICAM-1 expression and monocyte adhesion in the aortic root area. Furthermore, Il-6 mRNA expression was elevated in the aorta of OSM treated mice. CONCLUSION OSM induces endothelial activation in vitro in endothelial cells from different vascular beds through activation of the JAK/STAT cascade and in vivo in APOE*3Leiden.CETP mice. Since endothelial activation is an initial step in atherosclerosis development, OSM may play a role in the initiation of atherosclerotic lesion formation.
Collapse
Affiliation(s)
- Danielle van Keulen
- Laboratory of Experimental Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Haematology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Quorics B.V, Rotterdam, The Netherlands
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Marianne G. Pouwer
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Haematology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Alain J. van Gool
- TNO- Microbiology & Systems Biology, Zeist, The Netherlands
- Radboudumc, Nijmegen, The Netherlands
| | | | - Hans M. G. Princen
- TNO-Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Dennie Tempel
- Laboratory of Experimental Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Haematology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Quorics B.V, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
29
|
COMP-prohibitin 2 interaction maintains mitochondrial homeostasis and controls smooth muscle cell identity. Cell Death Dis 2018; 9:676. [PMID: 29867124 PMCID: PMC5986769 DOI: 10.1038/s41419-018-0703-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/06/2018] [Accepted: 05/14/2018] [Indexed: 12/22/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are highly phenotypically plastic, and loss of the contractile phenotype in VSMCs has been recognized at the early onset of the pathology of a variety of vascular diseases. However, the endogenous regulatory mechanism to maintain contractile phenotype in VSMCs remains elusive. Moreover, little has been known about the role of the mitochondrial bioenergetics in terms of VSMC homeostasis. Herein, we asked if glycoprotein COMP (Cartilage oligomeric matrix protein) is involved in mitochondrial bioenergetics and therefore regulates VSMCs homeostasis. By using fluorescence assay, subcellular western blot and liquid chromatography tandem mass spectrometry analysis, we found that extracellular matrix protein COMP unexpectedly localized within mitochondria. Further mitochondrial transplantation revealed that both mitochondrial and non-mitochondrial COMP maintained VSMC identity. Moreover, microarray analysis revealed that COMP deficiency impaired mitochondrial oxidative phosphorylation in VSMCs. Further study confirmed that COMP deficiency caused mitochondrial oxidative phosphorylation dysfunction accompanied by morphological abnormality. Moreover, the interactome of mitochondrial COMP revealed that COMP interacted with prohibitin 2, and COMP-prohibitin 2 interaction maintained mitochondrial homeostasis. Additionally, disruption of COMP-prohibitin 2 interaction caused VSMC dedifferentiation in vitro and enhanced the neointima formation post rat carotid artery injury in vivo. In conclusion, COMP-prohibitin 2 interaction in mitochondria plays an important role in maintaining the contractile phenotype of VSMCs by regulating mitochondrial oxidative phosphorylation. Maintaining the homeostasis of mitochondrial respiration through COMP-prohibitin 2 interaction may shed light on prevention of vascular disease.
Collapse
|
30
|
Lu QB, Wan MY, Wang PY, Zhang CX, Xu DY, Liao X, Sun HJ. Chicoric acid prevents PDGF-BB-induced VSMC dedifferentiation, proliferation and migration by suppressing ROS/NFκB/mTOR/P70S6K signaling cascade. Redox Biol 2017; 14:656-668. [PMID: 29175753 PMCID: PMC5716955 DOI: 10.1016/j.redox.2017.11.012] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 12/18/2022] Open
Abstract
Phenotypic switch of vascular smooth muscle cells (VSMCs) is characterized by increased expressions of VSMC synthetic markers and decreased levels of VSMC contractile markers, which is an important step for VSMC proliferation and migration during the development and progression of cardiovascular diseases including atherosclerosis. Chicoric acid (CA) is identified to exert powerful cardiovascular protective effects. However, little is known about the effects of CA on VSMC biology. Herein, in cultured VSMCs, we showed that pretreatment with CA dose-dependently suppressed platelet-derived growth factor type BB (PDGF-BB)-induced VSMC phenotypic alteration, proliferation and migration. Mechanistically, PDGF-BB-treated VSMCs exhibited higher mammalian target of rapamycin (mTOR) and P70S6K phosphorylation, which was attenuated by CA pretreatment, diphenyleneiodonium chloride (DPI), reactive oxygen species (ROS) scavenger N-acetyl-l-cysteine (NAC) and nuclear factor-κB (NFκB) inhibitor Bay117082. PDGF-BB-triggered ROS production and p65-NFκB activation were inhibited by CA. In addition, both NAC and DPI abolished PDGF-BB-evoked p65-NFκB nuclear translocation, phosphorylation and degradation of Inhibitor κBα (IκBα). Of note, blockade of ROS/NFκB/mTOR/P70S6K signaling cascade prevented PDGF-BB-evoked VSMC phenotypic transformation, proliferation and migration. CA treatment prevented intimal hyperplasia and vascular remodeling in rat models of carotid artery ligation in vivo. These results suggest that CA impedes PDGF-BB-induced VSMC phenotypic switching, proliferation, migration and neointima formation via inhibition of ROS/NFκB/mTOR/P70S6K signaling cascade. Chicoric acid attenuated PDGF-BB-evoked VSMC phenotypic transformation, proliferation and migration. Chicoric acid antagonized the activated ROS/NFκB/mTOR/P70S6K signaling pathway in VSMCs. Chicoric acid treatment prevented intimal hyperplasia in rat models of carotid artery ligation.
Collapse
Affiliation(s)
- Qing-Bo Lu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Ming-Yu Wan
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Pei-Yao Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Chen-Xing Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Dong-Yan Xu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Xiang Liao
- Department of Medical Imaging, General Hospital of Nanjing Military Area Command, Nanjing, Jiangsu 210002, PR China.
| | - Hai-Jian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
31
|
Sun HJ, Ren XS, Xiong XQ, Chen YZ, Zhao MX, Wang JJ, Zhou YB, Han Y, Chen Q, Li YH, Kang YM, Zhu GQ. NLRP3 inflammasome activation contributes to VSMC phenotypic transformation and proliferation in hypertension. Cell Death Dis 2017; 8:e3074. [PMID: 28981106 PMCID: PMC5680591 DOI: 10.1038/cddis.2017.470] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/17/2017] [Accepted: 08/22/2017] [Indexed: 01/12/2023]
Abstract
Inflammation is involved in pathogenesis of hypertension. NLRP3 inflammasome activation is a powerful mediator of inflammatory response via caspase-1 activation. The present study was designed to determine the roles and mechanisms of NLRP3 inflammasome in phenotypic modulation and proliferation of vascular smooth muscle cells (VSMCs) in hypertension. Experiments were conducted in spontaneously hypertensive rats (SHR) and primary aortic VSMCs. NLRP3 inflammasome activation was observed in the media of aorta in SHR and in the VSMCs from SHR. Knockdown of NLRP3 inhibited inflammasome activation, VSMC phenotypic transformation and proliferation in SHR-derived VSMCs. Increased NFκB activation, histone acetylation and histone acetyltransferase expression were observed in SHR-derived VSMCs and in media of aorta in SHR. Chromatin immunoprecipitation analysis revealed the increased histone acetylation, p65-NFκB and Pol II occupancy at the NLRP3 promoter in vivo and in vitro. Inhibition of NFκB with BAY11-7082 or inhibition of histone acetyltransferase with curcumin prevented the NLRP3 inflammasome activation, VSMC phenotype switching and proliferation in VSMCs from SHR. Moreover, curcumin repressed NFκB activation. Silencing of NLRP3 gene ameliorated hypertension, vascular remodeling, NLRP3 inflammasome activation and phenotype switching in the aorta of SHR. These results indicate that NLRP3 inflammasome activation response to histone acetylation and NFκB activation contributes to VSMC phenotype switching and proliferation and vascular remodeling in hypertension.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xing-Sheng Ren
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiao-Qing Xiong
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yun-Zhi Chen
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming-Xia Zhao
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jue-Jin Wang
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ye-Bo Zhou
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ying Han
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shanxi 710061, China
| | - Guo-Qing Zhu
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
32
|
Huhtinen A, Hongisto V, Laiho A, Löyttyniemi E, Pijnenburg D, Scheinin M. Gene expression profiles and signaling mechanisms in α 2B-adrenoceptor-evoked proliferation of vascular smooth muscle cells. BMC SYSTEMS BIOLOGY 2017; 11:65. [PMID: 28659168 PMCID: PMC5490158 DOI: 10.1186/s12918-017-0439-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND α2-adrenoceptors are important regulators of vascular tone and blood pressure. Regulation of cell proliferation is a less well investigated consequence of α2-adrenoceptor activation. We have previously shown that α2B-adrenoceptor activation stimulates proliferation of vascular smooth muscle cells (VSMCs). This may be important for blood vessel development and plasticity and for the pathology and therapeutics of cardiovascular disorders. The underlying cellular mechanisms have remained mostly unknown. This study explored pathways of regulation of gene expression and intracellular signaling related to α2B-adrenoceptor-evoked VSMC proliferation. RESULTS The cellular mechanisms and signaling pathways of α2B-adrenoceptor-evoked proliferation of VSMCs are complex and include redundancy. Functional enrichment analysis and pathway analysis identified differentially expressed genes associated with α2B-adrenoceptor-regulated VSMC proliferation. They included the upregulated genes Egr1, F3, Ptgs2 and Serpine1 and the downregulated genes Cx3cl1, Cav1, Rhoa, Nppb and Prrx1. The most highly upregulated gene, Lypd8, represents a novel finding in the VSMC context. Inhibitor library screening and kinase activity profiling were applied to identify kinases in the involved signaling pathways. Putative upstream kinases identified by two different screens included PKC, Raf-1, Src, the MAP kinases p38 and JNK and the receptor tyrosine kinases EGFR and HGF/HGFR. As a novel finding, the Src family kinase Lyn was also identified as a putative upstream kinase. CONCLUSIONS α2B-adrenoceptors may mediate their pro-proliferative effects in VSMCs by promoting the activity of bFGF and PDGF and the growth factor receptors EGFR, HGFR and VEGFR-1/2. The Src family kinase Lyn was also identified as a putative upstream kinase. Lyn is known to be expressed in VSMCs and has been identified as an important regulator of GPCR trafficking and GPCR effects on cell proliferation. Identified Ser/Thr kinases included several PKC isoforms and the β-adrenoceptor kinases 1 and 2. Cross-talk between the signaling mechanisms involved in α2B-adrenoceptor-evoked VSMC proliferation thus appears to involve PKC activation, subsequent changes in gene expression, transactivation of EGFR, and modulation of kinase activities and growth factor-mediated signaling. While many of the identified individual signals were relatively small in terms of effect size, many of them were validated by combining pathway analysis and our integrated screening approach.
Collapse
Affiliation(s)
- Anna Huhtinen
- Department of Pharmacology, Drug Development and Therapeutics, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Vesa Hongisto
- Toxicology Division, Misvik Biology Oy, Turku, Finland
| | - Asta Laiho
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Dirk Pijnenburg
- PamGene International BV, Wolvenhoek 10, 5211HH s’Hertogenbosch, The Netherlands
| | - Mika Scheinin
- Department of Pharmacology, Drug Development and Therapeutics, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
33
|
Liu H, Lin D, Xiang H, Chen W, Zhao S, Peng H, Yang J, Chen P, Chen S, Lu H. The role of tumor necrosis factor-like weak inducer of apoptosis in atherosclerosis via its two different receptors. Exp Ther Med 2017; 14:891-897. [PMID: 28781615 DOI: 10.3892/etm.2017.4600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
At present, it is commonly accepted that atherosclerosis is a chronic inflammatory disease characterized by disorder of the arterial wall. As one of the inflammatory cytokines of the tumor necrosis factor superfamily, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) participates in the formation and progression of atherosclerosis. TWEAK, when binding to its initial receptor, fibroblast growth factor inducible molecule 14 (Fn14), exerts adverse biological functions in atherosclerosis, including dysfunction of endothelial cells, phenotypic change of smooth muscle cells and inflammatory responses of monocytes/macrophages. However, accumulating data supports that, besides Fn14, TWEAK also binds to cluster of differentiation (CD)163, an anti-inflammatory cytokine and a scavenger receptor exclusively expressed by monocytes and macrophages. Furthermore, it has been demonstrated that CD163 is able to internalize TWEAK and likely elicits protective effects in atherosclerosis by terminating inflammation induced by TWEAK. In the present study, the role of TWEAK in atherosclerosis was reviewed, with a predominant focus on CD163 and Fn14 receptors.
Collapse
Affiliation(s)
- Hengdao Liu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Dan Lin
- Qingdao Center for Disease Control and Prevention, Qingdao, Shandong 266033, P.R. China
| | - Hong Xiang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Wei Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shaoli Zhao
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hui Peng
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jie Yang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Pan Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shuhua Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hongwei Lu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
34
|
Pan S, Lin H, Luo H, Gao F, Meng L, Zhou C, Jiang C, Guo Y, Ji Z, Chi J, Guo H. Folic acid inhibits dedifferentiation of PDGF-BB-induced vascular smooth muscle cells by suppressing mTOR/P70S6K signaling. Am J Transl Res 2017; 9:1307-1316. [PMID: 28386356 PMCID: PMC5376021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/07/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE Folic acid (FA) supplementation reduces the risk of atherosclerosis and stroke. Phenotypic change from differentiated to dedifferentiated vascular smooth muscle cells (VSMCs) plays an important role in atherosclerosis development; however, the exact mechanisms remain unknown. This study aimed to assess whether FA through mammalian target of rapamycin (mTOR)/P70S6K signaling inhibits platelet derived growth factor (PDGF-BB) induced VSMC dedifferentiation. METHODS VSMCs from primary cultures were identified by morphological observation and α-smooth muscle actin (α-SM-actin, α-SMA) immunocytochemistry. Then, VSMCs were induced by PDGF-BB and treated with varying FA concentrations. Rapamycin and MHY-1485 were used to inhibit or activate the mTOR/P70S6K pathway, respectively. Next, MTT, Transwell, and wound healing assays were employed to assess proliferation and migration of VSMCs. In addition, Western blotting was used to evaluate protein levels of α-SMA, calponin, osteopontin, mTOR, p-mTOR, P70S6K and p-P70S6K in VSMCs. RESULTS VSMCs showed phenotypic alteration from differentiated to dedifferentiated cells in response to PDGF-BB. MTT, Transwell and wound healing assays showed that FA markedly inhibited proliferation and migration in PDGF-BB-induced VSMCs, in a time and concentration-dependent manner. FA treatment increased the expression levels of the contractile phenotype marker proteins α-SMA and calponin compared with VSMCs stimulated by PDGF-BB alone. Furthermore, FA significantly suppressed mTOR and P70S6K phosphorylation compared with PDGF-BB alone. Similar to FA, downregulation of mTOR signaling by rapamycin inhibited VSMC dedifferentiation. In contrast, upregulation of mTOR signaling by MHY-1485 reversed the FA-induced inhibition of VSMC dedifferentiation. CONCLUSION Folic acid inhibits dedifferentiation of PDGF-BB-induced VSMCs by suppressing mTOR/P70S6K signaling.
Collapse
Affiliation(s)
- Sunlei Pan
- The First Clinical Medical College, Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Hui Lin
- The First Clinical Medical College, Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
| | - Hangqi Luo
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Feidan Gao
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Liping Meng
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Changzuan Zhou
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Chengjian Jiang
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Yan Guo
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Zheng Ji
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Jufang Chi
- The First Clinical Medical College, Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| | - Hangyuan Guo
- The First Clinical Medical College, Wenzhou Medical UniversityWenzhou 325000, Zhejiang, China
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang, China
| |
Collapse
|
35
|
Jiang D, Yang Y, Li D. Lipopolysaccharide induced vascular smooth muscle cells proliferation: A new potential therapeutic target for proliferative vascular diseases. Cell Prolif 2017; 50. [PMID: 28150467 DOI: 10.1111/cpr.12332] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) proliferation is involved in vascular atherosclerosis and restenosis. Recent studies have demonstrated that lipopolysaccharide (LPS) promotes VSMCs proliferation, but the signalling pathways which are involved are not completely understood. The purpose of this review was to summarize the existing knowledge of the role and molecular mechanisms involved in controlling VSMCs proliferation stimulated by LPS and mediated by toll-like receptor 4 (TLR4) signalling pathways. Moreover, the potential inhibitors of TLR4 signalling for VSMCs proliferation in proliferative vascular diseases are discussed.
Collapse
Affiliation(s)
- Dehua Jiang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Yang
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
36
|
Chen PY, Qin L, Li G, Tellides G, Simons M. Fibroblast growth factor (FGF) signaling regulates transforming growth factor beta (TGFβ)-dependent smooth muscle cell phenotype modulation. Sci Rep 2016; 6:33407. [PMID: 27634335 PMCID: PMC5025753 DOI: 10.1038/srep33407] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022] Open
Abstract
Smooth muscle cells (SMCs) in normal blood vessels exist in a highly differentiate state characterized by expression of SMC-specific contractile proteins ("contractile phenotype"). Following blood vessel injury in vivo or when cultured in vitro in the presence of multiple growth factors, SMC undergo a phenotype switch characterized by the loss of contractile markers and appearance of expression of non-muscle proteins ("proliferative phenotype"). While a number of factors have been reported to modulate this process, its regulation remains uncertain. Here we show that induction of SMC FGF signaling inhibits TGFβ signaling and converts contractile SMCs to the proliferative phenotype. Conversely, inhibition of SMC FGF signaling induces TGFβ signaling converting proliferating SMCs to the contractile phenotype, even in the presence of various growth factors in vitro or vascular injury in vivo. The importance of this signaling cross-talk is supported by in vivo data that show that an SMC deletion of a pan-FGF receptor adaptor Frs2α (fibroblast growth factor receptor substrate 2 alpha) in mice profoundly reduces neointima formation and vascular remodelling following carotid artery ligation. These results demonstrate that FGF-TGFβ signaling antagonism is the primary regulator of the SMC phenotype switch. Manipulation of this cross-talk may be an effective strategy for treatment of SMC-proliferation related diseases.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Vascular Surgery, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, China
| | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
37
|
Functional and Proteomic Investigations Reveal Major Royal Jelly Protein 1 Associated with Anti-hypertension Activity in Mouse Vascular Smooth Muscle Cells. Sci Rep 2016; 6:30230. [PMID: 27444336 PMCID: PMC4957218 DOI: 10.1038/srep30230] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/01/2016] [Indexed: 12/14/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are a major cell type of the arterial wall and their functionality is associated with blood pressure regulation. Although royal jelly (RJ) has reported effects on anti-hypertension, the mechanism of blood pressure regulation by major royal jelly protein 1 (MRJP1), the most abundant RJ protein, is still unknown. The mrjp1 gene was inserted into mouse VSMCs to investigate how MRJP1 influences VSMC functionality by functional and proteomic analysis. The expression of MRJP1 in VSMCs significantly reduced cell contraction, migration, and proliferation, suggesting a potential role in decreasing hypertension via action on VSMCs. These anti-hypertension activities were further observed in the changes of the proteome setting of mouse VSMCs. Among 675 different proteins after MRJP1 expression, 646 were down-regulated and significantly enriched in pathways implicated in VSMC contraction and migration, which suggest MRJP1 lowers VSMC contraction and migration by inhibiting muscle filament movement. The down-regulated proteins also enriched pathways in proliferation, indicating that MRJP1 hinders VSMC proliferation by reducing the supply of energy and genetic material. This is the first report integrating MRJP1 into VSMC, revealing the function and mechanism correlated with anti-hypertensive activity. This offers a therapeutic potential to control hypertension by gene-therapy using bee-products.
Collapse
|
38
|
Chen PY, Qin L, Li G, Tellides G, Simons M. Smooth muscle FGF/TGFβ cross talk regulates atherosclerosis progression. EMBO Mol Med 2016; 8:712-28. [PMID: 27189169 PMCID: PMC4931287 DOI: 10.15252/emmm.201506181] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The conversion of vascular smooth muscle cells (SMCs) from contractile to proliferative phenotype is thought to play an important role in atherosclerosis. However, the contribution of this process to plaque growth has never been fully defined. In this study, we show that activation of SMC TGFβ signaling, achieved by suppression of SMC fibroblast growth factor (FGF) signaling input, induces their conversion to a contractile phenotype and dramatically reduces atherosclerotic plaque size. The FGF/TGFβ signaling cross talk was observed in vitro and in vivo In vitro, inhibition of FGF signaling increased TGFβ activity, thereby promoting smooth muscle differentiation and decreasing proliferation. In vivo, smooth muscle-specific knockout of an FGF receptor adaptor Frs2α led to a profound inhibition of atherosclerotic plaque growth when these animals were crossed on Apoe(-/-) background and subjected to a high-fat diet. In particular, there was a significant reduction in plaque cellularity, increase in fibrous cap area, and decrease in necrotic core size. In agreement with these findings, examination of human coronary arteries with various degrees of atherosclerosis revealed a strong correlation between the activation of FGF signaling, loss of TGFβ activity, and increased disease severity. These results identify SMC FGF/TGFβ signaling cross talk as an important regulator of SMC phenotype switch and document a major contribution of medial SMC proliferation to atherosclerotic plaque growth.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, China
| | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Simons
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
39
|
Sun HJ, Zhao MX, Ren XS, Liu TY, Chen Q, Li YH, Kang YM, Wang JJ, Zhu GQ. Salusin-β Promotes Vascular Smooth Muscle Cell Migration and Intimal Hyperplasia After Vascular Injury via ROS/NFκB/MMP-9 Pathway. Antioxid Redox Signal 2016; 24:1045-57. [PMID: 26952533 DOI: 10.1089/ars.2015.6475] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS Media-to-intima migration of vascular smooth muscle cells (VSMCs) is critical to intimal thickening in atherosclerosis and restenosis after coronary angioplasty. The aim of this study is to determine the effects of salusin-β on VSMC migration and intimal hyperplasia after vascular injury and the underlying mechanism. RESULTS In vitro, salusin-β promoted VSMC migration, which was attenuated by matrix metalloproteinase (MMP)-9 inhibition. Inhibition or knockdown of p65-nuclear factor kappa beta (NFκB) in VSMCs suppressed salusin-β-induced MMP-9 expression and VSMC migration. Salusin-β increased NADPH oxidase 2 (NOX2) expression and reactive oxygen species (ROS) production, which were prevented by NOX2-small interfering RNA (siRNA) transfection. Salusin-β-induced p65-NFκB translocation, MMP-9 expression, and VSMC migration were inhibited by ROS scavenger, NADPH oxidase inhibitor, or NOX2-siRNA. In vivo, carotid artery ligation-induced vascular injury resulted in intimal hyperplasia in injured artery in rats. Salusin-β was upregulated in the injured carotid arteries of rats, which was attributed to reduced miR-133a-3p expression. Knockdown of salusin-β with siRNA attenuated the vascular injury-induced intimal thickening, p65-NFκB nuclear translocation, and NOX2 and MMP-9 expressions in rats. INNOVATION Salusin-β is a critical modulator in VSMC migration and neointima formation in response to vascular injury. CONCLUSIONS Salusin-β promotes VSMC migration and vascular injury-induced intimal hyperplasia via MMP-9 accumulation due to NOX2 activation, followed by ROS production, IκBα phosphorylation and degradation, and p65-NFκB translocation. We propose that salusin-β may be important in the VSMC migration and neointima of some vascular diseases. Antioxid. Redox Signal. 24, 1045-1057.
Collapse
Affiliation(s)
- Hai-Jian Sun
- 1 Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University , Nanjing, China
| | - Ming-Xia Zhao
- 1 Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University , Nanjing, China
| | - Xing-Sheng Ren
- 1 Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University , Nanjing, China
| | - Tong-Yan Liu
- 1 Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University , Nanjing, China
| | - Qi Chen
- 2 Department of Pathophysiology, Nanjing Medical University , Nanjing, China
| | - Yue-Hua Li
- 2 Department of Pathophysiology, Nanjing Medical University , Nanjing, China
| | - Yu-Ming Kang
- 3 Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine , Xi'an, China
| | - Jue-Jin Wang
- 1 Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University , Nanjing, China
| | - Guo-Qing Zhu
- 1 Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University , Nanjing, China
| |
Collapse
|
40
|
Hien TT, Turczyńska KM, Dahan D, Ekman M, Grossi M, Sjögren J, Nilsson J, Braun T, Boettger T, Garcia-Vaz E, Stenkula K, Swärd K, Gomez MF, Albinsson S. Elevated Glucose Levels Promote Contractile and Cytoskeletal Gene Expression in Vascular Smooth Muscle via Rho/Protein Kinase C and Actin Polymerization. J Biol Chem 2016; 291:3552-68. [PMID: 26683376 PMCID: PMC4751395 DOI: 10.1074/jbc.m115.654384] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 12/17/2015] [Indexed: 12/22/2022] Open
Abstract
Both type 1 and type 2 diabetes are associated with increased risk of cardiovascular disease. This is in part attributed to the effects of hyperglycemia on vascular endothelial and smooth muscle cells, but the underlying mechanisms are not fully understood. In diabetic animal models, hyperglycemia results in hypercontractility of vascular smooth muscle possibly due to increased activation of Rho-kinase. The aim of the present study was to investigate the regulation of contractile smooth muscle markers by glucose and to determine the signaling pathways that are activated by hyperglycemia in smooth muscle cells. Microarray, quantitative PCR, and Western blot analyses revealed that both mRNA and protein expression of contractile smooth muscle markers were increased in isolated smooth muscle cells cultured under high compared with low glucose conditions. This effect was also observed in hyperglycemic Akita mice and in diabetic patients. Elevated glucose activated the protein kinase C and Rho/Rho-kinase signaling pathways and stimulated actin polymerization. Glucose-induced expression of contractile smooth muscle markers in cultured cells could be partially or completely repressed by inhibitors of advanced glycation end products, L-type calcium channels, protein kinase C, Rho-kinase, actin polymerization, and myocardin-related transcription factors. Furthermore, genetic ablation of the miR-143/145 cluster prevented the effects of glucose on smooth muscle marker expression. In conclusion, these data demonstrate a possible link between hyperglycemia and vascular disease states associated with smooth muscle contractility.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/pathology
- Aged
- Animals
- Atherosclerosis/enzymology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cells, Cultured
- Contractile Proteins/agonists
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cytoskeletal Proteins/agonists
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 2/complications
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Gene Expression Regulation
- Humans
- Male
- Mice, Knockout
- Mice, Mutant Strains
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Protein Kinase C/chemistry
- Protein Kinase C/metabolism
- Signal Transduction
- rho GTP-Binding Proteins/agonists
- rho GTP-Binding Proteins/metabolism
- rho-Associated Kinases/chemistry
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Tran Thi Hien
- From the Departments of Experimental Medical Sciences and
| | | | - Diana Dahan
- From the Departments of Experimental Medical Sciences and
| | - Mari Ekman
- From the Departments of Experimental Medical Sciences and
| | - Mario Grossi
- From the Departments of Experimental Medical Sciences and
| | - Johan Sjögren
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Johan Nilsson
- Clinical Sciences, Lund University, BMC D12, SE-221 84 Lund, Sweden and
| | - Thomas Braun
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Thomas Boettger
- the Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany, and
| | - Eliana Garcia-Vaz
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | - Karin Stenkula
- From the Departments of Experimental Medical Sciences and
| | - Karl Swärd
- From the Departments of Experimental Medical Sciences and
| | - Maria F Gomez
- the Department of Clinical Sciences in Malmö, Lund University, 205 02 Malmö, Sweden
| | | |
Collapse
|
41
|
Zhu SB, Zhu J, Zhou ZZ, Xi EP, Wang RP, Zhang Y. TGF-β1 induces human aortic vascular smooth muscle cell phenotype switch through PI3K/AKT/ID2 signaling. Am J Transl Res 2015; 7:2764-2774. [PMID: 26885273 PMCID: PMC4731673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 12/12/2015] [Indexed: 06/05/2023]
Abstract
The vascular smooth muscle cell (VSMC) phenotypic switch is considered to be the key pathophysiological change in various cardiovascular diseases, such as aortic dissection, atherosclerosis, and hypertension. The results in this study showed that TGF-β1 promotes the proliferation, migration and morphological changes of VSMC.TGF-β1 promoted the expressions of PI3K, P-PI3K, AKT, P-AKT, ID2, and OPN protein and suppressed the expressions of α-SMA and SM22α protein; the opposite results were observed for TGF-β1 inhibitor group, AKT inhibitor group and Combined inhibitors group. After the stimulation of TGF-β1 signaling, the mRNA levels of PI3K, AKT, ID2, and OPN were the highest, while the mRNA levels of α-SMA and SM22α were the lowest; the opposite results were found in the same groups above. These results suggested the PI3K/AKT/ID2 signaling pathway is involved in TGF-β1-mediated human aortic VSMC phenotypic switching, that is from a contractile to synthetic phenotype, and Combined inhibitors was more effective in inhibiting the phenotypic switch than a single inhibitor. The Combined inhibitors experiments may provide new avenues for the prevention and treatment of thoracic aortic dissection (TAD) that are based on the pathological effects of phenotypic switching.
Collapse
Affiliation(s)
- Shui-Bo Zhu
- Department of Thoracic Cardiovascular Surgery, Wuhan General Hospital of Guangzhou Command Wuhan, 430070, People's Republic of China
| | - Jian Zhu
- Department of Thoracic Cardiovascular Surgery, Wuhan General Hospital of Guangzhou Command Wuhan, 430070, People's Republic of China
| | - Zi-Zi Zhou
- Department of Thoracic Cardiovascular Surgery, Wuhan General Hospital of Guangzhou Command Wuhan, 430070, People's Republic of China
| | - Er-Ping Xi
- Department of Thoracic Cardiovascular Surgery, Wuhan General Hospital of Guangzhou Command Wuhan, 430070, People's Republic of China
| | - Rong-Ping Wang
- Department of Thoracic Cardiovascular Surgery, Wuhan General Hospital of Guangzhou Command Wuhan, 430070, People's Republic of China
| | - Yu Zhang
- Department of Thoracic Cardiovascular Surgery, Wuhan General Hospital of Guangzhou Command Wuhan, 430070, People's Republic of China
| |
Collapse
|
42
|
Ke B, Zhang A, Wu X, Fang X. The Role of Krüppel-like Factor 4 in Renal Fibrosis. Front Physiol 2015; 6:327. [PMID: 26617530 PMCID: PMC4641914 DOI: 10.3389/fphys.2015.00327] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/29/2015] [Indexed: 12/22/2022] Open
Abstract
Chronic kidney disease (CKD) caused by renal fibrosis is an important public health concern. It is therefore necessary to understand the molecular pathogenesis of renal fibrosis in order to develop novel therapeutic strategies. KLF4 is the most extensively studied factor among the various members of the Krüppel-like factor (KLF) family of zinc finger-containing transcription factors. Many studies have demonstrated that KLF4 inhibits the activation of myofibroblasts and exerts an inhibitory effect on fibrosis. However, other studies have indicated that KLF4 may promote renal fibrosis. These controversial results suggest that KLF4 may be crucially involved in the development of renal fibrosis, although the underlying mechanism(s) remain unclear. Here, we summarize the recent progress made in understanding the role of KLF4 in renal fibrosis. Together, these findings suggest that KLF4 may participate in the development of renal fibrosis, but that its inhibition of fibrosis is greater than its promotion of the condition, which suggests that KLF4 may serve as a novel therapeutic target for renal fibrosis.
Collapse
Affiliation(s)
- Ben Ke
- Department of Nephrology, Nanchang University School of Medicine, Second Affiliated Hospital to Nanchang University Nanchang, China
| | - Afei Zhang
- Department of Nephrology, Nanchang University School of Medicine, Second Affiliated Hospital to Nanchang University Nanchang, China
| | - Xianfeng Wu
- Department of Nephrology, Nanchang University School of Medicine, Second Affiliated Hospital to Nanchang University Nanchang, China
| | - Xiangdong Fang
- Department of Nephrology, Nanchang University School of Medicine, Second Affiliated Hospital to Nanchang University Nanchang, China
| |
Collapse
|
43
|
Wu Y, Zhou J, Wang H, Wu Y, Gao Q, Wang L, Zhao Q, Liu P, Gao S, Wen W, Zhang W, Liu Y, Yuan Z. The activation of p38 MAPK limits the abnormal proliferation of vascular smooth muscle cells induced by high sodium concentrations. Int J Mol Med 2015; 37:74-82. [PMID: 26530729 PMCID: PMC4687433 DOI: 10.3892/ijmm.2015.2394] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 10/23/2015] [Indexed: 02/05/2023] Open
Abstract
The aim of the present study was to ascertain whether high sodium levels can directly promote the proliferation of vascular smooth muscle cells (VSMCs) and to elucidate the underlying mechanisms. Additional sodium chloride (NaCl) was added to the routine culture medium. Cell proliferation was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and 5-ethynyl-2′-deoxyuridine (EdU) incorporation assay. The mRNA expression level of proliferating cell nuclear antigen (PCNA) was measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The protein expression levels of PCNA and phosphorylated c-Jun amino N-terminal kinase (p-JNK), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) and phosphorylated p38 mitogen-activated protein kinase (p-p38 MAPK) were measured by western blot analysis. Cell proliferation assay revealed that Na+ rather than Cl− or osmotic pressure promoted the proliferation of the VSMCs. The high sodium level upregulated the expression of PCNA and the phosphorylation levels of JNK, ERK1/2 and p38 MAPK. The inhibition of JNK and ERK1/2 decreased PCNA expression. Of note, the inhibition of p38 MAPK using the inhibitor, SB203580, increased PCNA expression. However, when p38 MAPK was activated by anisomycin, PCNA expression was decreased. On the whole, our findings demonstrate that a relatively high sodium level per se directly promotes the proliferation of VSMCs through the JNK/ERK1/2/PCNA pathway. At the same time, this induction of the proliferation of VSMCs due to high sodium levels can be maintained at a low level via the activation of p38 MAPK.
Collapse
Affiliation(s)
- Yan Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Juan Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Huan Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yue Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qiyue Gao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lijun Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qiang Zhao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Peining Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shanshan Gao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wen Wen
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Weiping Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
44
|
Environmental carcinogens and mutational pathways in atherosclerosis. Int J Hyg Environ Health 2015; 218:293-312. [DOI: 10.1016/j.ijheh.2015.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/26/2015] [Accepted: 01/29/2015] [Indexed: 02/07/2023]
|
45
|
Mousavi SJ, Hamdy Doweidar M. Role of Mechanical Cues in Cell Differentiation and Proliferation: A 3D Numerical Model. PLoS One 2015; 10:e0124529. [PMID: 25933372 PMCID: PMC4416758 DOI: 10.1371/journal.pone.0124529] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/16/2015] [Indexed: 12/28/2022] Open
Abstract
Cell differentiation, proliferation and migration are essential processes in tissue regeneration. Experimental evidence confirms that cell differentiation or proliferation can be regulated according to the extracellular matrix stiffness. For instance, mesenchymal stem cells (MSCs) can differentiate to neuroblast, chondrocyte or osteoblast within matrices mimicking the stiffness of their native substrate. However, the precise mechanisms by which the substrate stiffness governs cell differentiation or proliferation are not well known. Therefore, a mechano-sensing computational model is here developed to elucidate how substrate stiffness regulates cell differentiation and/or proliferation during cell migration. In agreement with experimental observations, it is assumed that internal deformation of the cell (a mechanical signal) together with the cell maturation state directly coordinates cell differentiation and/or proliferation. Our findings indicate that MSC differentiation to neurogenic, chondrogenic or osteogenic lineage specifications occurs within soft (0.1-1 kPa), intermediate (20-25 kPa) or hard (30-45 kPa) substrates, respectively. These results are consistent with well-known experimental observations. Remarkably, when a MSC differentiate to a compatible phenotype, the average net traction force depends on the substrate stiffness in such a way that it might increase in intermediate and hard substrates but it would reduce in a soft matrix. However, in all cases the average net traction force considerably increases at the instant of cell proliferation because of cell-cell interaction. Moreover cell differentiation and proliferation accelerate with increasing substrate stiffness due to the decrease in the cell maturation time. Thus, the model provides insights to explain the hypothesis that substrate stiffness plays a key role in regulating cell fate during mechanotaxis.
Collapse
Affiliation(s)
- Seyed Jamaleddin Mousavi
- Group of Structural Mechanics and Materials Modeling (GEMM), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - Mohamed Hamdy Doweidar
- Group of Structural Mechanics and Materials Modeling (GEMM), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
- * E-mail:
| |
Collapse
|
46
|
Zhang L, Yu F, Wang L, Zheng J, Du Y, Huang Y, Liu B, Wang X, Kong W. ADAMTS-7 promotes vascular smooth muscle cells proliferation in vitro and in vivo. SCIENCE CHINA-LIFE SCIENCES 2015; 58:674-81. [PMID: 25921940 DOI: 10.1007/s11427-015-4843-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/11/2015] [Indexed: 10/23/2022]
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration are pivotal for the pathogenesis of atherosclerosis and post-angioplasty restenosis. We have recently reported that a disintegrin and metalloproteinase with thrombospondin motifs-7 (ADAMTS-7), a novel metalloproteinase, contributes directly to neointima formation by mediating VSMC migration. However, whether ADAMTS-7 affects VSMC proliferation remains unclear. In this study, we found that luminal adenoviral delivery of ADAMTS-7 aggravated intimal hyperplasia 7 d after injury, paralleled by an increased percentage of PCNA-positive cells in both intima and media. In contrast, perivascular administration of ADAMTS-7 siRNA, but not scrambled siRNA to injured arteries attenuated intimal thickening at day 7, paralleled with reduced intimal VSMC replication, without alteration of VSMC proliferation in the media. In accordance, [(3)H]-thymidine incorporation assay in primary cultured rat VSMCs revealed an enhanced replication rate (by 61%) upon ADAMTS-7 overexpression and retarded proliferation (by 23%) upon ADAMTS-7 siRNA administration. Our data demonstrates that ADAMTS-7 promotes VSMC proliferation both in vitro and in vivo. ADAMTS-7 may therefore serve as a novel therapeutic target for atherosclerosis and post-angioplasty restenosis.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Shi N, Chen SY. From nerve to blood vessel: a new role of Olfm2 in smooth muscle differentiation from human embryonic stem cell-derived mesenchymal cells. J Biomed Res 2015; 29:261-3. [PMID: 26243513 PMCID: PMC4547375 DOI: 10.7555/jbr.29.20150027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 03/08/2015] [Indexed: 01/10/2023] Open
Affiliation(s)
- Ning Shi
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|