51
|
Jin YH, Kim SA. 2-Methoxycinnamaldehyde inhibits the TNF-α-induced proliferation and migration of human aortic smooth muscle cells. Int J Mol Med 2016; 39:191-198. [PMID: 27922672 DOI: 10.3892/ijmm.2016.2818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 11/24/2016] [Indexed: 11/06/2022] Open
Abstract
The abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) is a crucial event in the development of atherosclerosis, and tumor necrosis factor-α (TNF-α) is actively involved in this process by enhancing the proliferation and migration of VSMCs. 2-Methoxycinnamaldehyde (MCA) is a natural compound of Cinnamomum cassia. Although 2-hydroxycinnamaldehyde (HCA), another compound from Cinnamomum cassia, has been widely studied with regard to its antitumor activity, MCA has not attracted researchers' interest due to its mild toxic effects on cancer cells and its mechanisms of action remain unknown. In this study, we examined the effects of MCA on the TNF-α-induced proliferation and migration of human aortic smooth muscle cells (HASMCs). As shown by our results, MCA inhibited TNF-α-induced cell proliferation by reducing the levels of cyclin D1, cyclin D3, CDK4 and CDK6, and increasing the levels of the cyclin-dependent kinase inhibitors, p21 and p27, without resulting in cellular cytotoxicity. Furthermore, MCA decreased the level of secreted matrix metalloproteinase (MMP)-9 by inhibiting MMP-9 transcription. Unexpectedly, MCA did not affect the TNF-α-induced levels of mitogen-activated protein kinases (MAPKs). However, by showing that MCA potently inhibited the degradation of IκBα and the subsequent nuclear translocation of nuclear factor-κB (NF-κB), we demonstrated that MCA exerts its effects through the NF-κB signaling pathway. MCA also effectively inhibited platelet-derived growth factor (PDGF)-induced HASMC migration. Taken together, these observations suggest that MCA has the potential for use as an anti-atherosclerotic agent.
Collapse
Affiliation(s)
- Young-Hee Jin
- Department of Biochemistry, Dongguk University College of Oriental Medicine, Gyeongju 780-714, Republic of Korea
| | - Soo-A Kim
- Department of Biochemistry, Dongguk University College of Oriental Medicine, Gyeongju 780-714, Republic of Korea
| |
Collapse
|
52
|
Beneit N, Fernández-García CE, Martín-Ventura JL, Perdomo L, Escribano Ó, Michel JB, García-Gómez G, Fernández S, Díaz-Castroverde S, Egido J, Gómez-Hernández A, Benito M. Expression of insulin receptor (IR) A and B isoforms, IGF-IR, and IR/IGF-IR hybrid receptors in vascular smooth muscle cells and their role in cell migration in atherosclerosis. Cardiovasc Diabetol 2016; 15:161. [PMID: 27905925 PMCID: PMC5134076 DOI: 10.1186/s12933-016-0477-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/22/2016] [Indexed: 01/02/2023] Open
Abstract
Background Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) is a major contributor to the development of atherosclerotic process. In a previous work, we demonstrated that the insulin receptor isoform A (IRA) and its association with the insulin-like growth factor-I receptor (IGF-IR) confer a proliferative advantage to VSMCs. However, the role of IR and IGF-IR in VSMC migration remains poorly understood. Methods Wound healing assays were performed in VSMCs bearing IR (IRLoxP+/+ VSMCs), or not (IR−/− VSMCs), expressing IRA (IRA VSMCs) or expressing IRB (IRB VSMCs). To study the role of IR isoforms and IGF-IR in experimental atherosclerosis, we used ApoE−/− mice at 8, 12, 18 and 24 weeks of age. Finally, we analyzed the mRNA expression of total IR, IRB isoform, IGF-IR and IGFs by qRT-PCR in the medial layer of human aortas. Results IGF-I strongly induced migration of the four cell lines through IGF-IR. In contrast, insulin and IGF-II only caused a significant increase of IRA VSMC migration which might be favored by the formation of IRA/IGF-IR receptors. Additionally, a specific IGF-IR inhibitor, picropodophyllin, completely abolished insulin- and IGF-II-induced migration in IRB, but not in IRA VSMCs. A significant increase of IRA and IGF-IR, and VSMC migration were observed in fibrous plaques from 24-week-old ApoE−/− mice. Finally, we observed a marked increase of IGF-IR, IGF-I and IGF-II in media from fatty streaks as compared with both healthy aortas and fibrolipidic lesions, favoring the ability of medial VSMCs to migrate into the intima. Conclusions Our data suggest that overexpression of IGF-IR or IRA isoform, as homodimers or as part of IRA/IGF-IR hybrid receptors, confers a stronger migratory capability to VSMCs as might occur in early stages of atherosclerotic process. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0477-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- N Beneit
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - C E Fernández-García
- Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - J L Martín-Ventura
- Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - L Perdomo
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Ó Escribano
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - J B Michel
- Inserm, U698, Universite Paris 7, CHU X-Bichat, Paris, France
| | - G García-Gómez
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - S Fernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - S Díaz-Castroverde
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - J Egido
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain.,Vascular Research Lab, IIS-Fundación Jiménez Diaz-Autonoma University, Madrid, Spain
| | - A Gómez-Hernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain. .,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain. .,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain.
| | - M Benito
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| |
Collapse
|
53
|
Saleh Al-Shehabi T, Iratni R, Eid AH. Anti-atherosclerotic plants which modulate the phenotype of vascular smooth muscle cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1068-1081. [PMID: 26776961 DOI: 10.1016/j.phymed.2015.10.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/27/2015] [Accepted: 10/30/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) remains the leading cause of global death, with atherosclerosis being a major contributor to this mortality. Several mechanisms are implicated in the pathogenesis of this disease. A key element in the development and progression of atherosclerotic lesions is the phenotype of vascular smooth muscle cells. Under pathophysiologic conditions such as injury, these cells switch from a contractile to a synthetic phenotype that often possesses high proliferative and migratory capacities. PURPOSE Despite major advances made in the management and treatment of atherosclerosis, mortality associated with this disease remains high. This mandates that other approaches be sought. Herbal medicine, especially for the treatment of CVD, has been gaining more attention in recent years. This is in no small part due to the evidence-based values associated with the consumption of many plants as well as the relatively cheaper prices, easier access and conventional folk medicine "inherited" over generations. Sections: In this review, we provide a brief introduction about the pathogenesis of atherosclerosis then we highlight the role of vascular smooth muscle cells in this disease, especially when a phenotypic switch of these cells arises. We then thoroughly discuss the various plants that show potentially beneficial effects as anti-atherosclerotic, with prime attention given to herbs and plants that inhibit the phenotypic switch of vascular smooth muscle cells. CONCLUSION Accumulating evidence provides the justification for the use of botanicals in the treatment or prevention of atherosclerosis. However, further studies, especially clinical ones, are warranted to better define several pharmacological parameters of these herbs, such as toxicity, tolerability, and efficacy.
Collapse
Affiliation(s)
- Tuqa Saleh Al-Shehabi
- Department of Health Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar.
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates.
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, PO Box 11-0236, Beirut, Lebanon ; Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar.
| |
Collapse
|
54
|
Daniel JM, Reichel CA, Schmidt-Woell T, Dutzmann J, Zuchtriegel G, Krombach F, Herold J, Bauersachs J, Sedding DG, Kanse SM. Factor VII-activating protease deficiency promotes neointima formation by enhancing leukocyte accumulation. J Thromb Haemost 2016; 14:2058-2067. [PMID: 27431088 DOI: 10.1111/jth.13417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 07/01/2016] [Indexed: 01/08/2023]
Abstract
Essentials Factor VII-activating protease (FSAP) is a plasma protease involved in vascular processes. Neointima formation was investigated after vascular injury in FSAP-/- mice. The neointimal lesion size and the accumulation of macrophages were increased in FSAP-/- mice. This was due to an increased activity of the chemokine (C-C motif) ligand 2 (CCL2). SUMMARY Background Factor VII-activating protease (FSAP) is a multifunctional circulating plasma serine protease involved in thrombosis and vascular remodeling processes. The Marburg I single-nucleotide polymorphism (MI-SNP) in the FSAP-coding gene is characterized by low proteolytic activity, and is associated with increased rates of stroke and carotid stenosis in humans. Objectives To determine whether neointima formation after vascular injury is increased in FSAP-/- mice. Methods and Results The neointimal lesion size and the proliferation of vascular smooth muscle cells (VSMCs) were significantly enhanced in FSAP-/- mice as compared with C57BL/6 control mice after wire-induced injury of the femoral artery. Accumulation of leukocytes and macrophages was increased within the lesions of FSAP-/- mice at day 3 and day 14. Quantitative zymography demonstrated enhanced activity of gelatinases/matrix metalloproteinase (MMP)-2 and MMP-9 within the neointimal lesions of FSAP-/- mice, and immunohistochemistry showed particular costaining of MMP-9 with accumulating leukocytes. Using intravital microscopy, we observed that FSAP deficiency promoted the intravascular adherence and the subsequent transmigration of leukocytes in vivo in response to chemokine ligand 2 (CCL2). CCL2 expression was increased in FSAP-/- monocytes but not in the vessel wall. There was no difference in the expression of platelet-derived growth factor (PDGF-BB). Conclusions FSAP deficiency causes an increase in CCL2 expression and CCL2-mediated infiltration of leukocytes into the injured vessel, thereby promoting SMC proliferation and migration by the activation of leukocyte-derived gelatinases. These results provide a possible explanation for the observed association of the loss-of-function MI-SNP with vascular proliferative diseases.
Collapse
Affiliation(s)
- J-M Daniel
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - C A Reichel
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - T Schmidt-Woell
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - J Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - G Zuchtriegel
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - F Krombach
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - J Herold
- Department of Cardiology, Angiology and Pneumology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - D G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - S M Kanse
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany.
- Oslo University Hospital and University of Oslo, Oslo, Norway.
| |
Collapse
|
55
|
Bodewes TCF, Johnson JM, Auster M, Huynh C, Muralidharan S, Contreras M, LoGerfo FW, Pradhan-Nabzdyk L. Intraluminal delivery of thrombospondin-2 small interfering RNA inhibits the vascular response to injury in a rat carotid balloon angioplasty model. FASEB J 2016; 31:109-119. [PMID: 27671229 DOI: 10.1096/fj.201600501r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/16/2016] [Indexed: 01/06/2023]
Abstract
In an effort to inhibit the response to vascular injury that leads to intimal hyperplasia, this study investigated the in vivo efficacy of intraluminal delivery of thrombospondin-2 (TSP-2) small interfering RNA (siRNA). Common carotid artery (CCA) balloon angioplasty injury was performed in rats. Immediately after denudation, CCA was transfected intraluminally (15 min) with one of the following: polyethylenimine (PEI)+TSP-2 siRNA, saline, PEI only, or PEI+control siRNA. CCA was analyzed at 24 h or 21 d by using quantitative real-time PCR and immunohistochemistry. TSP-2 gene and protein expression were significantly up-regulated after endothelial denudation at 24 h and 21 d compared with contralateral untreated, nondenuded CCA. Treatment with PEI+TSP-2 siRNA significantly suppressed TSP-2 gene expression (3.1-fold) at 24 h and TSP-2 protein expression, cell proliferation, and collagen deposition up to 21 d. These changes could be attributed to changes in TGF-β and matrix metalloproteinase-9, the downstream effectors of TSP-2. TSP-2 knockdown induced anti-inflammatory M2 macrophage polarization at 21 d; however, it did not significantly affect intima/media ratios. In summary, these data demonstrate effective siRNA transfection of the injured arterial wall and provide a clinically effective and translationally applicable therapeutic strategy that involves nonviral siRNA delivery to ameliorate the response to vascular injury.-Bodewes, T. C. F., Johnson, J. M., Auster, M., Huynh, C., Muralidharan, S., Contreras, M., LoGerfo, F. W., Pradhan-Nabzdyk, L. Intraluminal delivery of thrombospondin-2 small interfering RNA inhibits the vascular response to injury in a rat carotid balloon angioplasty model.
Collapse
Affiliation(s)
- Thomas C F Bodewes
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Vascular Surgery, University Medical Center, Utrecht, The Netherlands; and
| | - Joel M Johnson
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Auster
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Cindy Huynh
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| | - Sriya Muralidharan
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mauricio Contreras
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Frank W LoGerfo
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Leena Pradhan-Nabzdyk
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
56
|
Matrix Metalloproteinases in Non-Neoplastic Disorders. Int J Mol Sci 2016; 17:ijms17071178. [PMID: 27455234 PMCID: PMC4964549 DOI: 10.3390/ijms17071178] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/16/2016] [Accepted: 07/04/2016] [Indexed: 12/23/2022] Open
Abstract
The matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases belonging to the metzincin superfamily. There are at least 23 members of MMPs ever reported in human, and they and their substrates are widely expressed in many tissues. Recent growing evidence has established that MMP not only can degrade a variety of components of extracellular matrix, but also can cleave and activate various non-matrix proteins, including cytokines, chemokines and growth factors, contributing to both physiological and pathological processes. In normal conditions, MMP expression and activity are tightly regulated via interactions between their activators and inhibitors. Imbalance among these factors, however, results in dysregulated MMP activity, which causes tissue destruction and functional alteration or local inflammation, leading to the development of diverse diseases, such as cardiovascular disease, arthritis, neurodegenerative disease, as well as cancer. This article focuses on the accumulated evidence supporting a wide range of roles of MMPs in various non-neoplastic diseases and provides an outlook on the therapeutic potential of inhibiting MMP action.
Collapse
|
57
|
Cui D, Li H, Lei L, Chen C, Yan F. Nonsurgical periodontal treatment reduced aortic inflammation in ApoE(-/-) mice with periodontitis. SPRINGERPLUS 2016; 5:940. [PMID: 27386384 PMCID: PMC4929118 DOI: 10.1186/s40064-016-2637-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/21/2016] [Indexed: 01/07/2023]
Abstract
Background Although the correlation between periodontal infection and atherosclerotic lesions has been well recognized, whether and how the nonsurgical periodontal treatment (NSPT) can improve the vascular inflammation has not been investigated clearly. Methods Thirty-two apolipoprotein E−/− (apoE−/−) mice were randomly divided into four groups: (1) Con group: no treatment, blank control group; (2) Lig group: ligature-induced-periodontitis group; (3) Lig-N group: ligatures were removed on the 7th day; (4) Lig-SRP group: ligatures were removed on the 7th day, and scaling and root planing (SRP) were performed on the 9th day. All the animals were euthanized on the 30th day. Alveolar bone loss (ABL) was assessed under microcomputed tomography. Systemic inflammatory status and lipid contents in the plasma were detected. Expression of several surrogate markers for vascular inflammation was evaluated by immunohistology and quantitative real time PCR. Results NSPT reduced ABL, improved lipid profile, and inhibited systemic inflammation with reduced plasma interleukin-6 (IL-6) level in apoE−/− mice; in addition, reduced inflammation in arterial wall was observed in NSPT treated mice, showing less vascular cell adhesion molecule-1 expression and less macrophage adhesion; furthermore, NSPT improved elastic fiber fragmentation disorder in the aortic wall, thus preserved elasticity of aortic artery. Conclusion Ligature-induced periodontitis can lead to inflammatory response in the vascular wall and NSPT has beneficial effect on the early stage of atherosclerosis process in the articular wall by reducing systemic inflammation and improving lipid profile.
Collapse
Affiliation(s)
- Di Cui
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhong Yang Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Houxuan Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhong Yang Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Lang Lei
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhong Yang Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Changxing Chen
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhong Yang Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Fuhua Yan
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhong Yang Road, Nanjing, 210008 Jiangsu People's Republic of China
| |
Collapse
|
58
|
Chung YL, Pan CH, Wang CCN, Hsu KC, Sheu MJ, Chen HF, Wu CH. Methyl Protodioscin, a Steroidal Saponin, Inhibits Neointima Formation in Vitro and in Vivo. JOURNAL OF NATURAL PRODUCTS 2016; 79:1635-1644. [PMID: 27227546 DOI: 10.1021/acs.jnatprod.6b00217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Restenosis (or neointimal hyperplasia) remains a clinical limitation of percutaneous coronary angioplasty. Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are known to be involved in the development of restenosis. The present study aimed to investigate the ability and molecular mechanisms of methyl protodioscin (1), a steroidal saponin isolated from the root of Dioscorea nipponica, to inhibit neointimal formation. Our study demonstrated that 1 markedly inhibited the growth and migration of VSMCs (A7r5 cells). A cytometric analysis suggested that 1 induced growth inhibition by arresting VSMCs at the G1 phase of the cell cycle. A rat carotid artery balloon injury model indicated that neointima formation of the balloon-injured vessel was markedly reduced after extravascular administration of 1. Compound 1 decreased the expression levels of ADAM15 (a disintegrin and metalloprotease 15) and its downstream signaling pathways in the VSMCs. Moreover, the expressions and activities of matrix metalloproteinases (MMP-2 and MMP-9) were also suppressed by 1 in a concentration-dependent manner. Additionally, the molecular mechanisms appear to be mediated, in part, through the downregulation of ADAM15, FAK, ERK, and PI3K/Akt.
Collapse
MESH Headings
- ADAM Proteins/antagonists & inhibitors
- Algorithms
- Animals
- Aorta, Thoracic/cytology
- Carotid Artery Injuries
- Cell Movement
- Cell Proliferation
- Dioscorea/chemistry
- Diosgenin/analogs & derivatives
- Diosgenin/chemistry
- Diosgenin/pharmacology
- Dose-Response Relationship, Drug
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/isolation & purification
- Drugs, Chinese Herbal/pharmacology
- Hyperplasia/drug therapy
- Membrane Proteins/antagonists & inhibitors
- Models, Theoretical
- Molecular Structure
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Neointima/drug therapy
- Phosphatidylinositol 3-Kinases/metabolism
- Plant Roots/chemistry
- Rats
- Rats, Sprague-Dawley
- Saponins/chemistry
- Saponins/isolation & purification
- Saponins/pharmacology
- Signal Transduction
Collapse
Affiliation(s)
- Yun-Lung Chung
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan
| | - Chun-Hsu Pan
- Department of Pharmacy, Taipei Medical University , Taipei 11031, Taiwan
| | - Charles C-N Wang
- Department of Biomedical Informatics, Asia University , Taichung 41354, Taiwan
| | - Kai-Cheng Hsu
- Cancer Biology and Drug Dsicovery, Taipei Medical University , Taipei 11031, Taiwan
| | - Ming-Jyh Sheu
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan
| | - Hai-Feng Chen
- School of Pharmaceutical Sciences, Xiamen University , Xiamen 361005, China
| | - Chieh-Hsi Wu
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan
- Department of Pharmacy, Taipei Medical University , Taipei 11031, Taiwan
| |
Collapse
|
59
|
Gliesche DG, Hussner J, Witzigmann D, Porta F, Glatter T, Schmidt A, Huwyler J, Meyer Zu Schwabedissen HE. Secreted Matrix Metalloproteinase-9 of Proliferating Smooth Muscle Cells as a Trigger for Drug Release from Stent Surface Polymers in Coronary Arteries. Mol Pharm 2016; 13:2290-300. [PMID: 27241028 DOI: 10.1021/acs.molpharmaceut.6b00033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases are the leading causes of death in industrialized countries. Atherosclerotic coronary arteries are commonly treated with percutaneous transluminal coronary intervention followed by stent deployment. This treatment has significantly improved the clinical outcome. However, triggered vascular smooth muscle cell (SMC) proliferation leads to in-stent restenosis in bare metal stents. In addition, stent thrombosis is a severe side effect of drug eluting stents due to inhibition of endothelialization. The aim of this study was to develop and test a stent surface polymer, where cytotoxic drugs are covalently conjugated to the surface and released by proteases selectively secreted by proliferating smooth muscle cells. Resting and proliferating human coronary artery smooth muscle cells (HCASMC) and endothelial cells (HCAEC) were screened to identify an enzyme exclusively released by proliferating HCASMC. Expression analyses and enzyme activity assays verified selective and exclusive activity of the matrix metalloproteinase-9 (MMP-9) in proliferating HCASMC. The principle of drug release exclusively triggered by proliferating HCASMC was tested using the biodegradable stent surface polymer poly-l-lactic acid (PLLA) and the MMP-9 cleavable peptide linkers named SRL and AVR. The specific peptide cleavage by MMP-9 was verified by attachment of the model compound fluorescein. Fluorescein release was observed in the presence of MMP-9 secreting HCASMC but not of proliferating HCAEC. Our findings suggest that cytotoxic drug conjugated polymers can be designed to selectively release the attached compound triggered by MMP-9 secreting smooth muscle cells. This novel concept may be beneficial for stent endothelialization thereby reducing the risk of restenosis and thrombosis.
Collapse
Affiliation(s)
- Daniel G Gliesche
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel , 4056 Basel, Switzerland
| | - Janine Hussner
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel , 4056 Basel, Switzerland
| | - Dominik Witzigmann
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel , Basel 4056, Switzerland
| | - Fabiola Porta
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel , Basel 4056, Switzerland
| | - Timo Glatter
- Proteomics Core Facility, Biozentrum, University of Basel , Basel 4056, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel , Basel 4056, Switzerland
| | - Jörg Huwyler
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel , Basel 4056, Switzerland
| | | |
Collapse
|
60
|
Chen L, DeWispelaere A, Dastvan F, Osborne WRA, Blechner C, Windhorst S, Daum G. Smooth Muscle-Alpha Actin Inhibits Vascular Smooth Muscle Cell Proliferation and Migration by Inhibiting Rac1 Activity. PLoS One 2016; 11:e0155726. [PMID: 27176050 PMCID: PMC4866761 DOI: 10.1371/journal.pone.0155726] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/03/2016] [Indexed: 12/28/2022] Open
Abstract
Smooth muscle alpha-actin (SMA) is a marker for the contractile, non-proliferative phenotype of adult smooth muscle cells (SMCs). Upon arterial injury, expression of SMA and other structural proteins decreases and SMCs acquire a pro-migratory and proliferative phenotype. To what extent SMA regulates migration and proliferation of SMCs is unclear and putative signaling pathways involved remain to be elucidated. Here, we used lentiviral-mediated gene transfer and siRNA technology to manipulate expression of SMA in carotid mouse SMCs and studied effects of SMA. Overexpression of SMA results in decreased proliferation and migration and blunts serum-induced activation of the small GTPase Rac, but not RhoA. All inhibitory effects of SMA are rescued by expression of a constitutively active Rac1 mutant (V12rac1). Moreover, reduction of SMA expression by siRNA technology results in an increased activation of Rac. Taken together, this study identifies Rac1 as a downstream target for SMA to inhibit SMC proliferation and migration.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Surgery and Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States of America
| | - Allison DeWispelaere
- Department of Surgery and Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States of America
| | - Frank Dastvan
- Department of Surgery and Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States of America
| | - William R. A. Osborne
- Department of Pediatrics and Diabetes and Obesity Center of Excellence at the University of Washington, Seattle, WA, United States of America
| | - Christine Blechner
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Germany
| | - Guenter Daum
- Department of Surgery and Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
61
|
Yu YM, Chao TY, Chang WC, Chang MJ, Lee MF. Thymol reduces oxidative stress, aortic intimal thickening, and inflammation-related gene expression in hyperlipidemic rabbits. J Food Drug Anal 2016; 24:556-563. [PMID: 28911561 PMCID: PMC9336656 DOI: 10.1016/j.jfda.2016.02.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/19/2016] [Accepted: 02/15/2016] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis plays a key role in the development of cardiovascular diseases, and is often associated with oxidative stress and local inflammation. Thymol, a major polyphenolic compound in thyme, exhibits antioxidant and anti-inflammatory properties. In this study, we measured the in vitro antioxidant activity of thymol, and investigated the effect of thymol on high-fat-diet-induced hyperlipidemia and atherosclerosis. New Zealand white rabbits were fed with regular chow, high-fat and high-cholesterol diet (HC), T3, or T6 (HC with thymol supplementation at 3 mg/kg/d or 6 mg/kg/d, respectively) for 8 weeks. Aortic intimal thickening, serum lipid parameters, multiple inflammatory markers, proinflammatory cytokines, and atherosclerosis-associated indicators were significantly increased in the HC group but decreased upon thymol supplementation. In summary, thymol exhibits antioxidant activity, and may suppress the progression of high-fat-diet-induced hyperlipidemia and atherosclerosis by reducing aortic intimal lipid lesion, lowering serum lipids and oxidative stress, and alleviating inflammation-related responses.
Collapse
Affiliation(s)
- Ya-Mei Yu
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Tzu-Yu Chao
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Weng-Cheng Chang
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Margaret J Chang
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Ming-Fen Lee
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan.
| |
Collapse
|
62
|
Lee SJ, Won SY, Park SL, Song JH, Noh DH, Kim HM, Yin CS, Kim WJ, Moon SK. Rosa hybrida extract suppresses vascular smooth muscle cell responses by the targeting of signaling pathways, cell cycle regulation and matrix metalloproteinase-9 expression. Int J Mol Med 2016; 37:1119-26. [PMID: 26935151 DOI: 10.3892/ijmm.2016.2504] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 01/29/2016] [Indexed: 11/06/2022] Open
Abstract
The pharmacological effects of Rosa hybrida are well known in the cosmetics industry. However, the role of Rosa hybrida in cardiovascular biology had not previously been investigated, to the best of our knowledge. The aim of the present study was to elucidate the effect of water extract of Rosa hybrida (WERH) on platelet‑derived growth factor (PDGF)-stimulated vascular smooth muscle cells (VSMCs). VSMC proliferation, which was stimulated by PDGF, was inhibited in a non-toxic manner by WERH treatment, which also diminished the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and AKT. Treatment with WERH also induced G1-phase cell cycle arrest, which was due to the decreased expression of cyclins and cyclin-dependent kinases (CDKs), and induced p21WAF1 expression in PDGF-stimulated VSMCs. Moreover, WERH treatment suppressed the migration and invasion of VSMCs stimulated with PDGF. Treatment with WERH abolished the expression of matrix metalloproteinase-9 (MMP-9) and decreased the binding activity of nuclear factor-κB (NF-κB), activator protein-1 (AP-1), and specificity protein 1 (Sp1) motifs in PDGF-stimulated VSMCs. WERH treatment inhibited the proliferation of PDGF‑stimulated VSMCs through p21WAF1‑mediated G1-phase cell cycle arrest, by decreasing the kinase activity of cyclin/CDK complexes. Furthermore, WERH suppressed the PDGF-induced phosphorylation of ERK1/2 and AKT in VSMCs. Finally, treatment with WERH impeded the migration and invasion of VSMCs stimulated by PDGF by downregulating MMP-9 expression and a reduction in NF-κB, AP-1 and Sp1 activity. These results provide new insights into the effects of WERH on PDGF-stimulated VSMCs, and we suggest that WERH has the potential to act as a novel agent for the prevention and/or treatment of vascular diseases.
Collapse
Affiliation(s)
- Se-Jung Lee
- Department of Food and Nutrition, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Se Yeon Won
- Department of Food and Nutrition, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Sung Lyea Park
- Department of Food and Nutrition, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Jun-Hui Song
- Department of Food and Nutrition, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Dae-Hwa Noh
- Department of Food and Nutrition, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Hong-Man Kim
- Graduate School of East-West Medical Science, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Chang Shik Yin
- Acupuncture Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University College of Medicine, Cheongju, Chungbuk 361-763, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| |
Collapse
|
63
|
Zhang MJ, Zhou Y, Chen L, Wang X, Pi Y, Long CY, Sun MJ, Chen X, Gao CY, Li JC, Zhang LL. Impaired SIRT1 promotes the migration of vascular smooth muscle cell-derived foam cells. Histochem Cell Biol 2016; 146:33-43. [DOI: 10.1007/s00418-016-1408-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2016] [Indexed: 11/24/2022]
|
64
|
Alfakry H, Malle E, Koyani CN, Pussinen PJ, Sorsa T. Neutrophil proteolytic activation cascades: a possible mechanistic link between chronic periodontitis and coronary heart disease. Innate Immun 2016; 22:85-99. [PMID: 26608308 DOI: 10.1177/1753425915617521] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/13/2015] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular diseases are chronic inflammatory diseases that affect a large segment of society. Coronary heart disease (CHD), the most common cardiovascular disease, progresses over several years and affects millions of people worldwide. Chronic infections may contribute to the systemic inflammation and enhance the risk for CHD. Periodontitis is one of the most common chronic infections that affects up to 50% of the adult population. Under inflammatory conditions the activation of endogenous degradation pathways mediated by immune responses leads to the release of destructive cellular molecules from both resident and immigrant cells. Matrix metalloproteinases (MMPs) and their regulators can activate each other and play an important role in immune response via degrading extracellular matrix components and modulating cytokines and chemokines. The action of MMPs is required for immigrant cell recruitment at the site of inflammation. Stimulated neutrophils represent the major pathogen-fighting immune cells that upregulate expression of several proteinases and oxidative enzymes, which can degrade extracellular matrix components (e.g. MMP-8, MMP-9 and neutrophil elastase). The activity of MMPs is regulated by endogenous inhibitors and/or candidate MMPs (e.g. MMP-7). The balance between MMPs and their inhibitors is thought to mirror the proteolytic burden. Thus, neutrophil-derived biomarkers, including myeloperoxidase, may activate proteolytic destructive cascades that are involved in subsequent immune-pathological events associated with both periodontitis and CHD. Here, we review the existing studies on the contribution of MMPs and their regulators to the infection-related pathology. Also, we discuss the possible proteolytic involvement and role of neutrophil-derived enzymes as an etiological link between chronic periodontitis and CHD.
Collapse
Affiliation(s)
- Hatem Alfakry
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Chintan N Koyani
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Pirkko J Pussinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
65
|
Possible Mechanisms of Di(2-ethylhexyl) Phthalate-Induced MMP-2 and MMP-9 Expression in A7r5 Rat Vascular Smooth Muscle Cells. Int J Mol Sci 2015; 16:28800-11. [PMID: 26690114 PMCID: PMC4691078 DOI: 10.3390/ijms161226131] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 11/25/2022] Open
Abstract
Proliferation and migration of vascular smooth muscle cells (VSMC) are important in the development and/or progression of many cardiovascular diseases, including atherosclerosis. Evidence shows that matrix metalloproteinase (MMP)-2 and MMP-9 are related to the pathogenesis of atherosclerosis. The expressions of MMP-2 and MMP-9 in atherosclerosis are regulated via various pathways, such as p38 mitogen activated protein kinase (MAPK), extracellular signal regulated kinase 1 and 2 (ERK1/2), Akt, and nuclear factor kappa (NF-κB). Di(2-ethylhexyl) phthalate (DEHP) has been shown to induce atherosclerosis by increasing tumor necrosis factor (TNF)-α, interleukin (IL)-6, and intercellular adhesion molecule (ICAM) productions. However, whether DEHP poses any effects on MMP-2 or MMP-9 expression in VSMC has not yet been answered. In our studies, rat aorta VSMC was treated with DEHP (between 2 and 17.5 ppm) and p38 MAPK, ERK1/2, Akt, NF-κB, and MMP-2 and MMP-9 proteins and activities were measured. Results showed that the presence of DEHP can induce higher MMP-2 and MMP-9 expression than the controls. Similar results on MMP-regulating proteins, i.e., p38 MAPK, ERK1/2, Akt, and NF-κB, were also observed. In summary, our current results have showed that DEHP can be a potent inducer of atherosclerosis by increasing MMP-2 and MMP-9 expression at least through the regulations of p38 MAPK, ERK1/2, Akt, and NF-κB.
Collapse
|
66
|
The protective effect of bee venom on fibrosis causing inflammatory diseases. Toxins (Basel) 2015; 7:4758-72. [PMID: 26580653 PMCID: PMC4663532 DOI: 10.3390/toxins7114758] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/24/2015] [Accepted: 11/05/2015] [Indexed: 02/06/2023] Open
Abstract
Bee venom therapy is a treatment modality that may be thousands of years old and involves the application of live bee stings to the patient’s skin or, in more recent years, the injection of bee venom into the skin with a hypodermic needle. Studies have proven the effectiveness of bee venom in treating pathological conditions such as arthritis, pain and cancerous tumors. However, there has not been sufficient review to fully elucidate the cellular mechanisms of the anti-inflammatory effects of bee venom and its components. In this respect, the present study reviews current understanding of the mechanisms of the anti-inflammatory properties of bee venom and its components in the treatment of liver fibrosis, atherosclerosis and skin disease.
Collapse
|
67
|
p21WAF1 Is Required for Interleukin-16-Induced Migration and Invasion of Vascular Smooth Muscle Cells via the p38MAPK/Sp-1/MMP-9 Pathway. PLoS One 2015; 10:e0142153. [PMID: 26544695 PMCID: PMC4636239 DOI: 10.1371/journal.pone.0142153] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 10/19/2015] [Indexed: 01/30/2023] Open
Abstract
Interleukin-16 (IL-16) is a lymphocyte chemoattractant factor well known for its role in immune responses, but its role in vascular disease is unknown. Here, we explored the novel physiological function of IL-16 in vascular smooth muscle cells (VSMCs). The expression of IL-16 and its receptor CD4 was observed in VSMCs. Treatment with IL-16 enhanced the migration and invasion by VSMCs without altering the proliferative potential. IL-16 induced MMP-9 expression via the binding activity of transcription factors NF-κB, AP-1, and Sp-1 motifs in VSMCs. Among the relevant signaling pathways examined, only p38MAPK phosphorylation was significantly stimulated in IL-16-treated VSMCs. Treatment with p38MAPK inhibitor SB203580 prevented the IL-16-induced migration and invasion of VSMCs. SB203580 treatment inhibited the MMP-9 expression and activation of Sp-1 binding in IL-16-treated VSMCs, and siRNA knockdown of CD4 expression blocked the induction of migration, invasion, p38MAPK phosphorylation, MMP-9 expression, and Sp-1 binding activation stimulated by IL-16. The IL-16 induced cell-cycle-inhibitor p21WAF1 expression in VSMCs, but had no effect on the expression levels of other cell-cycle negative regulators. Finally, blockage of p21WAF1 function with specific siRNA abolished the IL-16-induced elevation of migration, invasion, p38MAPK phosphorylation, MMP-9 expression, and Sp-1 binding activation in VSMCs. Taken together, p21WAF1 was required for the induction of p38MAPK-mediated MMP-9 expression via activation of the Sp-1 binding motif, which led to migration and invasion of VSMCs interacting with IL-16/CD4. These results could provide that IL-16 is a new target in the treatment of vascular diseases such as atherosclerosis and re-stenosis.
Collapse
|
68
|
Unterman S, Freiman A, Beckerman M, Abraham E, Stanley JR, Levy E, Artzi N, Edelman E. Tuning of collagen scaffold properties modulates embedded endothelial cell regulatory phenotype in repair of vascular injuries in vivo. Adv Healthc Mater 2015; 4:2220-8. [PMID: 26333178 PMCID: PMC4664078 DOI: 10.1002/adhm.201500457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/31/2015] [Indexed: 01/08/2023]
Abstract
Perivascularly implanted matrix embedded endothelial cells (MEECs) are potent regulators of inflammation and intimal hyperplasia following vascular injuries. Endothelial cells (ECs) in collagen scaffolds adopt a reparative phenotype with significant therapeutic potential. Although the biology of MEECs is increasingly understood, tuning of scaffold properties to control cell-substrate interactions is less well-studied. It is hypothesized that modulating scaffold degradation would change EC phenotype. Scaffolds with differential degradation are prepared by cross-linking and predegradation. Vascular injury increases degradation and the presence of MEECs retards injury-mediated degradation. MEECs respond to differential scaffold properties with altered viability in vivo, suppressed smooth muscle cell (SMC) proliferation in vitro, and altered interleukin-6 and matrix metalloproteinase-9 expression. When implanted perivascularly to a murine carotid wire injury, tuned scaffolds change MEEC effects on vascular repair and inflammation. Live animal imaging enables real-time tracking of cell viability, inflammation, and scaffold degradation, affording an unprecedented understanding of interactions between cells, substrate, and tissue. MEEC-treated injuries improve endothelialization and reduce SMC hyperplasia over 14 d. These data demonstrate the potent role material design plays in tuning MEEC efficacy in vivo, with implications for the design of clinical therapies.
Collapse
Affiliation(s)
- Shimon Unterman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alina Freiman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ort Braude College, Karmiel, Israel
| | - Margarita Beckerman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ort Braude College, Karmiel, Israel
| | - Eytan Abraham
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James R.L. Stanley
- CBSET, Inc., Concord Biomedical Sciences and Emerging Technologies, Lexington, MA 02421, USA
| | - Ela Levy
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ort Braude College, Karmiel, Israel
| | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elazer Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
69
|
Freise C, Sommer K, Querfeld U. Protective effects of the polyphenols (+)-episesamin and sesamin against PDGF-BB-induced activation of vascular smooth muscle cells are mediated by induction of haem oxygenase-1 and inhibition of mitogenic signalling. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
70
|
Anti-Restenotic Roles of Dihydroaustrasulfone Alcohol Involved in Inhibiting PDGF-BB-Stimulated Proliferation and Migration of Vascular Smooth Muscle Cells. Mar Drugs 2015; 13:3046-60. [PMID: 25988521 PMCID: PMC4446617 DOI: 10.3390/md13053046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 05/03/2015] [Accepted: 05/05/2015] [Indexed: 01/23/2023] Open
Abstract
Dihydroaustrasulfone alcohol (DA), an active compound firstly isolated from marine corals, has been reported to reveal anti-cancer and anti-inflammation activities. These reported activities of DA raised a possible application in anti-restenosis. Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) and the stimulation of platelet-derived growth factor (PDGF)-BB play major pathological processes involved in the development of restenosis. Experimental results showed that DA markedly reduced balloon injury-induced neointima formation in the rat carotid artery model and significantly inhibited PDGF-BB-stimulated proliferation and migration of VSMCs. Our data further demonstrated that translational and active levels of several critical signaling cascades involved in VSMC proliferation, such as extracellular signal-regulated kinase/mitogen-activated protein kinases (ERK/MAPK), phosphatidylinositol 3-kinase (PI3K)/AKT, and signal transducer and activator of transcription (STAT), were obviously inhibited. In addition, DA also decreased the activation and expression levels of gelatinases (matrix metalloproteinase (MMP)-2 and MMP-9) involved in cell migration. In conclusion, our findings indicate that DA can reduce balloon injury-neointimal hyperplasia, the effect of which may be modulated through suppression of VSMC proliferation and migration. These results suggest that DA has potential application as an anti-restenotic agent for the prevention of restenosis.
Collapse
|
71
|
Phloretin Inhibits Platelet-derived Growth Factor-BB–induced Rat Aortic Smooth Muscle Cell Proliferation, Migration, and Neointimal Formation After Carotid Injury. J Cardiovasc Pharmacol 2015; 65:444-55. [DOI: 10.1097/fjc.0000000000000213] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
72
|
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
|
73
|
Zhang HJ, Wang J, Liu HF, Zhang XN, Zhan M, Chen FL. Overexpression of mimecan in human aortic smooth muscle cells inhibits cell proliferation and enhances apoptosis and migration. Exp Ther Med 2015; 10:187-192. [PMID: 26170933 DOI: 10.3892/etm.2015.2444] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 01/12/2015] [Indexed: 11/06/2022] Open
Abstract
The pathogenesis of atherosclerosis is multifactorial. The proliferation and migration of vascular smooth muscle cells (VSMCs) are significant in the genesis and development of atherosclerosis plaques, and the degradation of VSMCs plays a crucial role in the process. Mimecan is a member of the Keratan sulfate family of proteoglycans, which are leucine-rich proteoglycans. It has been demonstrated that mimecan is associated with arteriogenesis and atherosclerosis. In the present study, the effect of mimecan on the characteristics of cultured human aortic smooth muscle cells (HASMCs) was investigated. In vitro, human mimecan was stably overexpressed in HASMCs using a lentiviral system. It was observed that the proliferation rate of HASMCs transduced with mimecan was lower compared with that of control cells; overexpression of mimecan induced HASMC apoptosis. To determine the effect of mimecan on HASMC migration, a Transwell cell culture chamber and sterile cloning cylinder assays were used, and it was noted that mimecan enhanced the migration of HASMCs horizontally and vertically. These data indicated that mimecan may be involved in the pathogenesis of atherosclerosis by regulating the proliferation, apoptosis and migration of VSMCs.
Collapse
Affiliation(s)
- Hui-Jie Zhang
- Department of Endocrinology, Third People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - Jing Wang
- Department of Endocrinology, Third People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - Hui-Fang Liu
- Department of Endocrinology, Third People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| | - Xiao-Na Zhang
- Shanghai Institution of Endocrinology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Ming Zhan
- Shanghai Institution of Endocrinology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Feng-Ling Chen
- Department of Endocrinology, Third People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201900, P.R. China
| |
Collapse
|
74
|
Freise C, Querfeld U. The lignan (+)-episesamin interferes with TNF-α-induced activation of VSMC via diminished activation of NF-ĸB, ERK1/2 and AKT and decreased activity of gelatinases. Acta Physiol (Oxf) 2015; 213:642-52. [PMID: 25267105 DOI: 10.1111/apha.12400] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/03/2014] [Accepted: 09/24/2014] [Indexed: 12/15/2022]
Abstract
AIM Activation of vascular smooth muscle cells (VSMC), a key event in the pathogenesis of atherosclerosis, is triggered by inflammatory stimuli such as tumour necrosis factor-alpha (TNF-α) causing a mitogenic VSMC response. The polyphenol (+)-episesamin (ES) was shown to counteract TNF-α-induced effects, for example in macrophages. Aiming for novel therapeutic options, we here investigated whether ES protects VSMC from TNF-α-induced growth and migration, which both contribute to the onset and progression of atherosclerosis. METHODS Human and murine VSMC were treated with combinations of ES and TNF-α. Expressions of mRNA were analyzed by RT-PCR. Enzymatic activities and proliferation were determined by specific substrate assays. Cell signalling was analyzed by Western blot and reporter gene assays. Migration was assessed by wound healing assays. RESULTS ES at 1-10 μm reduced basal and TNF-α-induced VSMC proliferation and migration due to impaired activation of extracellular signal-regulated kinases (ERK)1/2, Akt (protein kinase B), nuclear factor-kappa B (NF-ĸB) and vascular cell adhesion molecule (VCAM)-1. This was accompanied by reduced expression and secretion of matrix metalloproteinases (MMP)-2/-9, which are known to promote VSMC migration. Specific inhibitors of Akt, NF-ĸB and MMP-2/-9 reduced TNF-α-induced VSMC proliferation, confirming ES-specific effects. Besides, ES reduced TNF-α- and H₂O₂ -induced oxidative stress and in parallel induces anti-inflammatory haem oxygenase (HO)-1 expression. CONCLUSION ES interferes with inflammation-associated VSMC activation and subsequent decreased proliferation and migration due to anti-oxidative properties and impaired activation of NF-ĸB, known contributors to atherogenesis. These results suggest ES as a complemental treatment of VSMC specific vascular diseases such as atherosclerosis.
Collapse
Affiliation(s)
- C. Freise
- Department of Pediatric Nephrology and Center for Cardiovascular Research; Charité - University Medicine; Campus Virchow Clinic; Berlin Germany
| | - U. Querfeld
- Department of Pediatric Nephrology; Charité - University Medicine; Campus Virchow Clinic; Berlin Germany
| |
Collapse
|
75
|
Lin ZW, Wang Z, Zhu GP, Li BW, Xie WL, Xiang DC. Hypertensive vascular remodeling was inhibited by Xuezhikang through the regulation of Fibulin-3 and MMPs in spontaneously hypertensive rats. Int J Clin Exp Med 2015; 8:2118-27. [PMID: 25932142 PMCID: PMC4402789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 01/25/2015] [Indexed: 12/08/2022]
Abstract
Fibulin-3, an extracellular glycoprotein, has been suggested as having functions in vessels. In hypertension, extracellular matrix, matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) play important roles in cardiovascular remodeling. However, the role of Fibulin-3 as an extracellular glycoprotein in hypertensive vascular remodeling remains unclear. Our study was to determine whether Fibulin-3 and TIMPs/MMPs would affect vascular structure during hypertension and the treatment of Xuezhikang. Thirty spontaneously hypertensive rats (SHRs) aged 8 weeks were randomized to three groups: SHRs control group (SHRs group, n=10), group treated with low dose Xuezhikang (XZK-L, 20 mg/kg/d, n=10) and group treated with high dose Xuezhikang (XZK-H, 200 mg/kg/d, n=10), the normal group was comprised of ten Wistar-Kyoto (WKY) rats of the same age. We showed that serum nitric oxide (NO) in control group was significantly lower than WKY group (P<0.05). Concomitantly, serum oxidized low-density lipoprotein (ox-LDL) was higher than WKY group (P<0.05). The treatment of high dose Xuezhikang significantly dicreased ox-LDL, left ventricular mass index (LVMI) and Wall-to-lumen area ratio (W/L) of thoracic aorta (P<0.05), while serum NO was significantly increasing (P<0.05). Moreover, the expressions of Fibulin-3 and MMP-2, 9 at both protein and mRNA levels were significantly higher in thoracic aorta of SHRs group compared to WKY group by immunohistochemistry and western blotting (P<0.05). However, the levels of Fibulin-3 and MMP-2, 9 were significantly decreased in XZK-H group compared to control group (P<0.05). The level of TIMP-3 had no significance difference between SHRs and WKY groups (P>0.05). So the levels of Fibulin-3 and MMP-2, 9 in SHRs could be inhibited by Xuezhikang. Furthermore, a strong correlation in transcript expression was established between Fibulin-3, and MMP-2 (r=0.81, P<0.05) and MMP-9 (r=0.92, P<0.05) through immunohistochemistry. In summary, the overexpression of Fibulin-3 and MMP-2, 9 levels were associated with hypertension and vascular remodeling and inhibited by Xuezhikang. Fibulin-3 is a candidate in the pathogenesis of cardiovascular remodeling in hypertension.
Collapse
Affiliation(s)
- Zhong-Wei Lin
- Graduate School of Southern Medical UniversityGuangzhou, China
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhou, China
| | - Zhuo Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China
| | - Gui-Ping Zhu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhou, China
| | - Bo-Wei Li
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhou, China
| | - Wen-Lin Xie
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China
| | - Ding-Cheng Xiang
- Department of Cardiology, Guangzhou General Hospital of Guangzhou Military CommandGuangzhou, China
| |
Collapse
|
76
|
Park SL, Won SY, Song JH, Kambe T, Nagao M, Kim WJ, Moon SK. EPO gene expression promotes proliferation, migration and invasion via the p38MAPK/AP-1/MMP-9 pathway by p21WAF1 expression in vascular smooth muscle cells. Cell Signal 2014; 27:470-8. [PMID: 25496832 DOI: 10.1016/j.cellsig.2014.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/15/2014] [Accepted: 12/01/2014] [Indexed: 11/15/2022]
Abstract
The use of recombinant human erythropoietin (rHuEpo) can lead to hypertrophy and hyperplasia, and has induced the proliferation of vascular smooth muscle cells (VSMCs). The effect of the EPO gene in the migration and invasion of VSMCs remains unclear. In this study, overexpression of the EPO gene increased the DNA synthesis and phosphorylation of ERK1/2 and p38MAPK in VSMCs. In addition, EPO gene expression induced the migration and invasion of VSMCs via the expression of MMP-9 by the activation of NF-κB and AP-1 binding. A blockade of p38MAPK by specific p38MAPK inhibitor SB203580 led to a suppression of the increased DNA synthesis, migration, and invasion of VSMCs that was induced by the EPO gene. SB203580 treatment blocked the increased expression of MMP-9 through the binding activity of AP-1. Transfection of the EPO gene with VSMCs was associated with the up-regulation of cyclin D1/CDK4, cyclin E/CDK2, and p21WAF1, and with the down-regulation of p27KIP1. The specific suppression of p21WAF1 expression by siRNA rescued the enhancement of DNA synthesis via the phosphorylation of p38MAPK and the increase in migration and invasion through AP-1-mediated MMP-9 expression in EPO gene transfectants. These novel findings demonstrate that p21WAF1 regulates the proliferation, migration and invasion of VSMC induced by EPO gene.
Collapse
Affiliation(s)
- Sung Lyea Park
- School of Food Science and Technology, Chung-Ang University, Ansung 456-756, Republic of Korea
| | - Se Yeon Won
- School of Food Science and Technology, Chung-Ang University, Ansung 456-756, Republic of Korea
| | - Jun-Hui Song
- School of Food Science and Technology, Chung-Ang University, Ansung 456-756, Republic of Korea
| | - Taiho Kambe
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University, Cheongju, Chungbuk 361-763, Republic of Korea
| | - Masaya Nagao
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University, Cheongju, Chungbuk 361-763, Republic of Korea
| | - Wun-Jae Kim
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Sung-Kwon Moon
- School of Food Science and Technology, Chung-Ang University, Ansung 456-756, Republic of Korea.
| |
Collapse
|
77
|
Favreau JT, Liu C, Yu P, Tao M, Mauro C, Gaudette GR, Ozaki CK. Acute reductions in mechanical wall strain precede the formation of intimal hyperplasia in a murine model of arterial occlusive disease. J Vasc Surg 2014; 60:1340-1347. [PMID: 24139980 PMCID: PMC3989476 DOI: 10.1016/j.jvs.2013.07.113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 07/22/2013] [Accepted: 07/27/2013] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Intimal hyperplasia (IH) continues to plague the durability of vascular interventions. Employing a validated murine model, ultrasound biomicroscopy, and speckle-tracking algorithms, we tested the hypothesis that reduced cyclic arterial wall strain results in accentuated arterial wall IH. METHODS A 9-0 suture was tied around the left mouse (n = 10) common carotid artery and a 35-gauge (outer diameter = 0.14 mm) blunt mandrel. We previously showed that mandrel removal results in a ∼78% reduction in diameter and ∼85% reduction in flow, with subsequent delayed induction of IH by day 28. Preoperative, postoperative day-4 (before measurable IH), and postoperative day-27 circumferential wall strains were measured in locations 1, 2, and 3 mm proximal to the stenosis and in the same locations on the contralateral (nonstenosed) carotid. At postoperative day 28, arteries were perfusion fixed and arterial wall morphology was assessed microscopically in the same regions. RESULTS Strains were the same in all locations preoperatively. Wall strain was decreased in all regions proximal to the stenosis by day 4 (0.26 ± 0.01 to 0.11 ± 0.02; P < .001), while strains remained unchanged for the contralateral artery (P = .45). No statistical regional differences in mean strain or IH were noted at any time point for the experimental or contralateral artery. Based on the median, regions were divided into those with low strain (≤0.1) and high strain (>0.1). Average preoperative strains in both groups were the same (0.27 ± 0.09 and 0.27 ± 0.08). All segments in the low-strain group (n = 13) demonstrated significant IH formation by day 28, while only 31% of the high strain group demonstrated any detectable IH at day 28. (Mean low-strain intimal thickness = 32 ± 20 μm, high strain = 8.0 ± 16 μm; P < .01). Changes in cross-sectional area at diastole drove the reduction in strain in the low-strain group, increasing significantly from preoperatively to day 4 (P = .04), while lumen cross-section at systole remained unchanged (P = .46). Cross-sectional area at diastole and systole in the high-strain group remained unchanged from preoperatively to day 4 (P = .67). CONCLUSIONS Early reduction in arterial wall strain is associated with subsequent development of hemodynamically induced IH.
Collapse
Affiliation(s)
- John T Favreau
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Mass; Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Mass
| | - Chengwei Liu
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Mass; Division of Vascular Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China
| | - Peng Yu
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Mass
| | - Ming Tao
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Mass
| | - Christine Mauro
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Mass
| | - Glenn R Gaudette
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Mass
| | - C Keith Ozaki
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital/Harvard Medical School, Boston, Mass.
| |
Collapse
|
78
|
Zhang SM, Zhu LH, Chen HZ, Zhang R, Zhang P, Jiang DS, Gao L, Tian S, Wang L, Zhang Y, Wang PX, Zhang XF, Zhang XD, Liu DP, Li H. Interferon regulatory factor 9 is critical for neointima formation following vascular injury. Nat Commun 2014; 5:5160. [PMID: 25319116 PMCID: PMC4218966 DOI: 10.1038/ncomms6160] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/05/2014] [Indexed: 12/28/2022] Open
Abstract
Interferon regulatory factor 9 (IRF9) has various biological functions and regulates cell survival; however, its role in vascular biology has not been explored. Here we demonstrate a critical role for IRF9 in mediating neointima formation following vascular injury. Notably, in mice, IRF9 ablation inhibits the proliferation and migration of vascular smooth muscle cells (VSMCs) and attenuates intimal thickening in response to injury, whereas IRF9 gain-of-function promotes VSMC proliferation and migration, which aggravates arterial narrowing. Mechanistically, we show that the transcription of the neointima formation modulator SIRT1 is directly inhibited by IRF9. Importantly, genetic manipulation of SIRT1 in smooth muscle cells or pharmacological modulation of SIRT1 activity largely reverses the neointima-forming effect of IRF9. Together, our findings suggest that IRF9 is a vascular injury-response molecule that promotes VSMC proliferation and implicate a hitherto unrecognized 'IRF9-SIRT1 axis' in vasculoproliferative pathology modulation.
Collapse
Affiliation(s)
- Shu-Min Zhang
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Li-Hua Zhu
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Ran Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Peng Zhang
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Ding-Sheng Jiang
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Lu Gao
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Song Tian
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Lang Wang
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Yan Zhang
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Pi-Xiao Wang
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Xiao-Fei Zhang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiao-Dong Zhang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Hongliang Li
- 1] Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China [2] Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| |
Collapse
|
79
|
Lemaire M, Lemarié CA, Flores Molina M, Guilbert C, Lehoux S, Mann KK. Genetic deletion of LXRα prevents arsenic-enhanced atherosclerosis, but not arsenic-altered plaque composition. Toxicol Sci 2014; 142:477-88. [PMID: 25273567 DOI: 10.1093/toxsci/kfu197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Arsenic exposure has been linked to an increased incidence of atherosclerosis. Previously, we have shown in vitro and in vivo that arsenic inhibits transcriptional activation of the liver X receptors (LXRs), key regulators of lipid homeostasis. Therefore, we evaluated the role of LXRα in arsenic-induced atherosclerosis using the apoE(-/-) mouse model. Indeed, deletion of LXRα protected apoE(-/-) mice against the proatherogenic effects of arsenic. We have previously shown that arsenic changes the plaque composition in apoE(-/-) mice. Arsenic decreased collagen content in the apoE(-/-) model, and we have observed the same diminution in LXRα(-/-)apoE(-/-) mice. However, the collagen-producing smooth muscle cells (SMCs) were decreased in apoE(-/-), but increased in LXRα(-/-)apoE(-/-). Although transcriptional activation of collagen remained the same in SMC from both genotypes, arsenic-exposed LXRα(-/-)apoE(-/-) plaques had increased matrix metalloproteinase activity compared with both control LXRα(-/-)apoE(-/-) and apoE(-/-), which could be responsible for both the decrease in plaque collagen and the SMC invasion. In addition, arsenic increased plaque lipid accumulation in both genotypes. However, macrophages, the cells known to retain lipid within the plaque, were unchanged in arsenic-exposed apoE(-/-) mice, but decreased in LXRα(-/-)apoE(-/-). We confirmed in vitro that these cells retained more lipid following arsenic exposure and are more sensitive to apoptosis than apoE(-/-). Mice lacking LXRα are resistant to arsenic-enhanced atherosclerosis, but arsenic-exposed LXRα(-/-)apoE(-/-) mice still present a different plaque composition pattern than the arsenic-exposed apoE(-/-) mice.
Collapse
Affiliation(s)
- Maryse Lemaire
- *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2 *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2
| | - Catherine A Lemarié
- *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2 *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2
| | - Manuel Flores Molina
- *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2
| | - Cynthia Guilbert
- *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2
| | - Stéphanie Lehoux
- *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2 *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2
| | - Koren K Mann
- *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2 *Department of Oncology, Lady Davis Institute for Medical Research, and Department of Medicine, McGill University, Montréal, Canada H3T 1E2
| |
Collapse
|
80
|
Ham SA, Lee H, Hwang JS, Kang ES, Yoo T, Paek KS, Do JT, Park C, Oh JW, Kim JH, Han CW, Seo HG. Activation of Peroxisome Proliferator-Activated Receptor δ Inhibits Angiotensin II-Induced Activation of Matrix Metalloproteinase-2 in Vascular Smooth Muscle Cells. J Vasc Res 2014; 51:221-30. [DOI: 10.1159/000365250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 06/07/2014] [Indexed: 11/19/2022] Open
|
81
|
Alizadeh AM, Shiri S, Farsinejad S. Metastasis review: from bench to bedside. Tumour Biol 2014; 35:8483-523. [PMID: 25104089 DOI: 10.1007/s13277-014-2421-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/29/2014] [Indexed: 12/19/2022] Open
Abstract
Cancer is the final result of uninhibited cell growth that involves an enormous group of associated diseases. One major aspect of cancer is when cells attack adjacent components of the body and spread to other organs, named metastasis, which is the major cause of cancer-related mortality. In developing this process, metastatic cells must successfully negotiate a series of complex steps, including dissociation, invasion, intravasation, extravasation, and dormancy regulated by various signaling pathways. In this review, we will focus on the recent studies and collect a comprehensive encyclopedia in molecular basis of metastasis, and then we will discuss some new potential therapeutics which target the metastasis pathways. Understanding the new aspects on molecular mechanisms and signaling pathways controlling tumor cell metastasis is critical for the development of therapeutic strategies for cancer patients that would be valuable for researchers in both fields of molecular and clinical oncology.
Collapse
Affiliation(s)
- Ali Mohammad Alizadeh
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, 1419733141, Iran,
| | | | | |
Collapse
|
82
|
PARK SUNGSOO, KIM WUNJAE, MOON SUNGKWON. Gleditsia sinensis thorn extract inhibits the proliferation and migration of PDGF-induced vascular smooth muscle cells. Mol Med Rep 2014; 10:2031-8. [DOI: 10.3892/mmr.2014.2422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 03/19/2014] [Indexed: 11/06/2022] Open
|
83
|
Johnson JL. Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. Cardiovasc Res 2014; 103:452-60. [PMID: 25053639 DOI: 10.1093/cvr/cvu171] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
After a period of relative senescence in the field of vascular smooth muscle cell (VSMC) research with particular regards to atherosclerosis, the last few years has witnessed a resurgence, with extensive research re-assessing potential molecular mechanisms and pathways that modulate VSMC behaviour within the atherosclerotic-prone vessel wall and the atherosclerotic plaque itself. Attention has focussed on the pathological contribution of VSMC in plaque calcification; systemic and local mediators such as inflammatory molecules and lipoproteins; autocrine and paracrine regulators which affect cell-cell and cell to matrix contacts alongside cytoskeletal changes. In this brief focused review, recent insights that have been gained into how a myriad of recently identified factors can influence the pathological behaviour of VSMC and their subsequent contribution to atherosclerotic plaque development and progression has been discussed. An overriding theme is the mechanisms involved in the alterations of VSMC function during atherosclerosis.
Collapse
Affiliation(s)
- Jason Lee Johnson
- Laboratory of Cardiovascular Pathology, School of Clinical Sciences, University of Bristol, Research Floor Level Seven, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| |
Collapse
|
84
|
Abstract
The extracellular matrix (ECM) is an essential component of the human body that is responsible for the proper function of various organs. Changes in the ECM have been implicated in the pathogenesis of several cardiovascular conditions including atherosclerosis, restenosis, and heart failure. Matrix components, such as collagens and noncollagenous proteins, influence the function and activity of vascular cells, particularly vascular smooth muscle cells and macrophages. Matrix proteins have been shown to be implicated in the development of atherosclerotic complications, such as plaque rupture, aneurysm formation, and calcification. ECM proteins control ECM remodeling through feedback signaling to matrix metalloproteinases (MMPs), which are the key players of ECM remodeling in both normal and pathological conditions. The production of MMPs is closely related to the development of an inflammatory response and is subjected to significant changes at different stages of atherosclerosis. Indeed, blood levels of circulating MMPs may be useful for the assessment of the inflammatory activity in atherosclerosis and the prediction of cardiovascular risk. The availability of a wide variety of low-molecular MMP inhibitors that can be conjugated with various labels provides a good perspective for specific targeting of MMPs and implementation of imaging techniques to visualize MMP activity in atherosclerotic plaques and, most interestingly, to monitor responses to antiatheroslerosis therapies. Finally, because of the crucial role of ECM in cardiovascular repair, the regenerative potential of ECM could be successfully used in constructing engineered scaffolds and vessels that mimic properties of the natural ECM and consist of the native ECM components or composite biomaterials. These scaffolds possess a great promise in vascular tissue engineering.
Collapse
|
85
|
Li W, Mata KM, Mazzuca MQ, Khalil RA. Altered matrix metalloproteinase-2 and -9 expression/activity links placental ischemia and anti-angiogenic sFlt-1 to uteroplacental and vascular remodeling and collagen deposition in hypertensive pregnancy. Biochem Pharmacol 2014; 89:370-85. [PMID: 24704473 DOI: 10.1016/j.bcp.2014.03.017] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 03/21/2014] [Accepted: 03/24/2014] [Indexed: 01/05/2023]
Abstract
Preeclampsia is a complication of pregnancy manifested as maternal hypertension and often fetal growth restriction. Placental ischemia could be an initiating event, but the linking mechanisms leading to hypertension and growth restriction are unclear. We have shown an upregulation of matrix metalloproteinases (MMPs) during normal pregnancy (Norm-Preg). To test the role of MMPs in hypertensive-pregnancy (HTN-Preg), maternal and fetal parameters, MMPs expression, activity and distribution, and collagen and elastin content were measured in uterus, placenta and aorta of Norm-Preg rats and in rat model of reduced uteroplacental perfusion pressure (RUPP). Maternal blood pressure was higher, and uterine, placental and aortic weight, and the litter size and pup weight were less in RUPP than Norm-Preg rats. Western blots and gelatin zymography revealed decreases in amount and gelatinase activity of MMP-2 and MMP-9 in uterus, placenta and aorta of RUPP compared with Norm-Preg rats. Immunohistochemistry confirmed reduced MMPs in uterus, placenta and aortic media of RUPP rats. Collagen, but not elastin, was more abundant in uterus, placenta and aorta of RUPP than Norm-Preg rats. The anti-angiogenic factor soluble fms-like tyrosine kinase-1 (sFlt-1) decreased MMPs in uterus, placenta and aorta of Norm-Preg rats, and vascular endothelial growth factor (VEGF) reversed the decreases in MMPs in tissues of RUPP rats. Thus placental ischemia and anti-angiogenic sFlt-1 decrease uterine, placental and vascular MMP-2 and MMP-9, leading to increased uteroplacental and vascular collagen, and growth-restrictive remodeling in HTN-Preg. Angiogenic factors and MMP activators may reverse the decrease in MMPs and enhance growth-permissive remodeling in preeclampsia.
Collapse
Affiliation(s)
- Wei Li
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Karina M Mata
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Marc Q Mazzuca
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
86
|
Min SK, Min SI, Jeong EM, Cho SY, Ha J, Kim SJ, Kim IG. Intimal hyperplasia in loop-injured carotid arteries is attenuated in transglutaminase 2-null mice. J Korean Med Sci 2014; 29:363-9. [PMID: 24616585 PMCID: PMC3945131 DOI: 10.3346/jkms.2014.29.3.363] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 12/10/2013] [Indexed: 01/25/2023] Open
Abstract
Arterial restenosis frequently develops after open or endovascular surgery due to intimal hyperplasia. Since tissue transglutaminase (TG2) is known to involve in fibrosis, wound healing, and extracellular matrix remodeling, we examined the role of TG2 in the process of intimal hyperplasia using TG2-null mice. The neointimal formation was compared between TG2-null and wild-type (C57BL/6) mice by two different injury models; carotid ligation and carotid loop injury. In ligation model, there was no difference in intimal thickness between two groups. In loop injury model, intimal hyperplasia developed in both groups and the intimal/medial area ratio was significantly reduced in TG2-null mice (P = 0.007). TG2 was intensely stained in neointimal cells in 2 weeks. In situ activity of TG2 in the injured arteries steadily increased until 4 weeks compared to uninjured arteries. Taken together, intimal hyperplasia was significantly reduced in TG2-null mice, indicating that TG2 has an important role in the development of intimal hyperplasia. This suggests that TG2 may be a novel target to prevent the arterial restenosis after vascular surgery.
Collapse
Affiliation(s)
- Seung-Kee Min
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Il Min
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Eui Man Jeong
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Yup Cho
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jongwon Ha
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Joon Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - In-Gyu Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
87
|
Wu W, Hu Y, Li J, Zhu W, Ha T, Que L, Liu L, Zhu Q, Chen Q, Xu Y, Li C, Li Y. Silencing of Pellino1 improves post-infarct cardiac dysfunction and attenuates left ventricular remodelling in mice. Cardiovasc Res 2014; 102:46-55. [DOI: 10.1093/cvr/cvu007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
88
|
Mittal B, Mishra A, Srivastava A, Kumar S, Garg N. Matrix metalloproteinases in coronary artery disease. Adv Clin Chem 2014; 64:1-72. [PMID: 24938016 DOI: 10.1016/b978-0-12-800263-6.00001-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Matrix metalloproteinases (MMP) are a family of zinc-containing endoproteinases that degrade extracellular matrix (ECM) components. MMP have important roles in the development, physiology and pathology of cardiovascular system. Metalloproteases also play key roles in adverse cardiovascular remodeling, atherosclerotic plaque formation and plaque instability, vascular smooth muscle cell (SMC) migration and restenosis that lead to coronary artery disease (CAD), and progressive heart failure. The study of MMP in developing animal model cardiovascular systems has been helpful in deciphering numerous pathologic conditions in humans. Increased peripheral blood MMP-2 and MMP-9 in acute coronary syndrome (ACS) may be useful as noninvasive tests for detection of plaque vulnerability. MMP function can be modulated by certain pharmacological drugs that can be exploited for treatment of ACS. CAD is a polygenic disease and hundreds of genes contribute toward its predisposition. A large number of sequence variations in MMP genes have been identified. Case-control association studies have highlighted their potential association with CAD and its clinical manifestations. Although results thus far are inconsistent, meta-analysis has demonstrated that MMP-3 Glu45Lys and MMP-9 1562C/T gene polymorphisms were associated with CAD risk.
Collapse
|
89
|
Interferon regulatory factor 8 modulates phenotypic switching of smooth muscle cells by regulating the activity of myocardin. Mol Cell Biol 2013; 34:400-14. [PMID: 24248596 DOI: 10.1128/mcb.01070-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Interferon regulatory factor 8 (IRF8), a member of the IRF transcription factor family, was recently implicated in vascular diseases. In the present study, using the mouse left carotid artery wire injury model, we unexpectedly observed that the expression of IRF8 was greatly enhanced in smooth muscle cells (SMCs) by injury. Compared with the wild-type controls, IRF8 global knockout mice exhibited reduced neointimal lesions and maintained SMC marker gene expression. We further generated SMC-specific IRF8 transgenic mice using an SM22α-driven IRF8 plasmid construct. In contrast to the knockout mice, mice with SMC-overexpressing IRF8 exhibited a synthetic phenotype and enhanced neointima formation. Mechanistically, IRF8 inhibited SMC marker gene expression through regulating serum response factor (SRF) transactivation in a myocardin-dependent manner. Furthermore, a coimmunoprecipitation assay indicated a direct interaction of IRF8 with myocardin, in which a specific region of myocardin was essential for recruiting acetyltransferase p300. Altogether, IRF8 is crucial in modulating SMC phenotype switching and neointima formation in response to vascular injury via direct interaction with the SRF/myocardin complex.
Collapse
|
90
|
Guo J, Li L, Wu YJ, Yan Y, Xu XN, Wang SB, Yuan TY, Fang LH, Du GH. Inhibitory Effects of Brazilin on the Vascular Smooth Muscle Cell Proliferation and Migration Induced by PDGF-BB. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2013; 41:1283-96. [DOI: 10.1142/s0192415x13500869] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Abnormal vascular smooth muscle cell (VSMC) proliferation and migration contribute to the pathogenesis of vascular diseases including atherosclerosis and restenosis. Brazilin isolated from the heartwood of Caesalpinia sappan L. has been reported to exhibit various biological activities, such as anti-platelet aggregation, anti-inflammation, vasorelaxation and pro-apoptosis. However, the functional effects of Brazilin on VSMCs remain unexplored. The present study investigated the potential effects of Brazilin on platelet-derived growth factor (PDGF)-BB induced VSMC proliferation and migration as well as the underlying mechanism of action. VSMC proliferation and migration were measured by Crystal Violet Staining, wound-healing and Boyden chamber assays, respectively. Cell cycle was analyzed by flow cytometry. Enzymatic action of matrix metalloproteinase-9 (MMP-9) was carried out by gelatin zymography. Expression of adhesion molecules, cell cycle regulatory proteins, the phosphorylated levels of PDGF receptor β (PDGF-Rβ), Src, extracellular signal regulated kinase (ERK) and Akt were tested by immunoblotting. The present study demonstrated that pretreatment with Brazilin dose-dependently inhibited PDGF-BB stimulated VSMC proliferation and migration, which were associated with a cell-cycle arrest at G0/G1 phase, a reduction in the adhesion molecule expression and MMP-9 activation in VSMCs. Furthermore, the increase in PDGF-Rβ, Src, ERK1/2 and Akt phosphorylation induced by PDGF-BB were suppressed by Brazilin. These findings indicate that Brazilin inhibits PDGF-BB induced VSMC proliferation and migration, and the inhibitory effects of Brazilin may be associated with the blockade of PDGF-Rβ - ERK1/2 and Akt signaling pathways. In conclusion, the present study implicates that Brazilin may be useful as an anti-proliferative agent for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| | - Li Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| | - Yu-Jie Wu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu Yan
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiao-Na Xu
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shou-Bao Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| | - Tian-Yi Yuan
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lian-Hua Fang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
| |
Collapse
|
91
|
Lei J, Vodovotz Y, Tzeng E, Billiar TR. Nitric oxide, a protective molecule in the cardiovascular system. Nitric Oxide 2013; 35:175-85. [DOI: 10.1016/j.niox.2013.09.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/02/2013] [Accepted: 09/24/2013] [Indexed: 12/19/2022]
|
92
|
Deatrick KB, Luke CE, Elfline MA, Sood V, Baldwin J, Upchurch GR, Jaffer FA, Wakefield TW, Henke PK. The effect of matrix metalloproteinase 2 and matrix metalloproteinase 2/9 deletion in experimental post-thrombotic vein wall remodeling. J Vasc Surg 2013; 58:1375-1384.e2. [PMID: 23490298 PMCID: PMC3688659 DOI: 10.1016/j.jvs.2012.11.088] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 11/14/2012] [Accepted: 11/21/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Vein wall fibrotic injury following deep venous thrombosis (VT) is associated with elevated matrix metalloproteinases (MMPs). Whether and by what mechanism MMP2 contributes to vein wall remodeling after VT is unknown. METHODS Stasis VT was produced by ligation of the inferior vena cava and tissue was harvested at 2, 8, and 21 days in MMP2 -/- and genetic wild type (WT) mice. Tissue analysis by immunohistochemistry, enzyme-linked immunosorbent assay, real-time polymerase chain reaction, and zymography was performed. RESULTS Thrombus resolution was less at 8 days in MMP2 -/- compared with WT, evidenced by a 51% increase in VT size (P < .01), and threefold fewer von Willebrand's factor positive channels (P < .05). In MMP2 -/- mice, the main phenotypic fibrotic differences occurred at 8 days post-VT, with significantly less vein wall collagen content (P = .013), fourfold lower procollagen III gene expression (P < .01), but no difference in procollagen I compared with WT. Decreased inflammation in MMP2 -/- vein walls was suggested by ∼ threefold reduced TNFα and IL-1β at 2 days and 8 days post-VT (P < .05). A fourfold increase in vein wall monocytes (P = .03) with threefold decreased apoptosis (P < .05), but no difference in cellular proliferation at 8 days was found in MMP2 -/- compared with WT. As increased compensatory MMP9 activity was observed in the MMP2 -/-mice, MMP2/9 double null mice had thrombus induced with VT harvest at 8 days. Consistently, twofold larger VT, a threefold decrease in vein wall collagen, and a threefold increase in monocytes were found (all P < .05). Similar findings were observed in MMP9 -/- mice administered an exogenous MMP2 inhibitor. CONCLUSIONS In stasis VT, deletion of MMP2 was associated with less midterm vein wall fibrosis and inflammation, despite an increase in monocytes. Consideration that VT resolution was impaired with MMP2 (and MMP2/9) deletion suggests direct inhibition will likely also require anticoagulant therapy.
Collapse
Affiliation(s)
- Kristopher B Deatrick
- Conrad Jobst Vascular Research Laboratory, Section of Vascular Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Xiao Q, Zhang F, Grassia G, Hu Y, Zhang Z, Xing Q, Yin X, Maddaluno M, Drung B, Schmidt B, Maffia P, Ialenti A, Mayr M, Xu Q, Ye S. Matrix metalloproteinase-8 promotes vascular smooth muscle cell proliferation and neointima formation. Arterioscler Thromb Vasc Biol 2013; 34:90-8. [PMID: 24158518 DOI: 10.1161/atvbaha.113.301418] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We investigated the role of matrix metalloproteinase-8 (MMP8) in neointima formation and in vascular smooth muscle cell (VSMC) migration and proliferation. APPROACH AND RESULTS After carotid artery wire injuring, MMP8(-/-)/apoE(-/-) mice had fewer proliferating cells in neointimal lesions and smaller lesion sizes. Ex vivo assays comparing VSMCs isolated from MMP8 knockout and wild-type mice showed that MMP8 knockout decreased proliferation and migration. Proteomics analysis revealed that a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) had lower concentrations in MMP8 knockout VSMC culture media than in MMP8 wild-type VSMC culture media. Western blot, flow cytometric, and immunocytochemical analyses showed that MMP8 knockout VSMCs contained more pro-ADAM10 but less mature ADAM10, more N-cadherin, and β-catenin in the plasma membrane but less β-catenin in the nucleus and less cyclin D1. Treatment of MMP8 wild-type VSMCs with an ADAM10 inhibitor, GI254023X, or siRNA knockdown of ADAM10 in MMP8 wild-type VSMCs inhibited proliferation and migration, increased N-cadherin and β-catenin in the plasma membrane, reduced β-catenin in the nucleus, and decreased cyclin D1 expression. Incubation of MMP8 knockout VSMCs with a recombinant ADAM10 rescued the proliferative and migratory ability of MMP8 knockout VSMCs and increased cyclin D1 expression. Furthermore, immunohistochemical analyses showed colocalization of ADAM10 with VSMCs and N-cadherin, and nuclear accumulation of β-catenin in the neointima in apoE(-/-)/MMP8(+/+) mice. CONCLUSIONS MMP8 enhances VSMC proliferation via an ADAM10, N-cadherin, and β-catenin-mediated pathway and plays an important role in neointima formation.
Collapse
Affiliation(s)
- Qingzhong Xiao
- From William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (Q. Xiao, F.Z., S.Y.); Department of Cardiology, Peking University People's Hospital, Beijing, China (F.Z.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (G.G., M. Maddaluno, P.M., A.I.); Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Y.H., Z.Z., Q. Xing, X.Y., M. Mayr, Q. Xu); Clemens Schöpf Institute, Technische Universität Darmstadt, Darmstadt, Germany (B.D., B.S.); Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Chen YF, Tsai HY, Wu KJ, Siao LR, Wood WG. Pipoxolan ameliorates cerebral ischemia via inhibition of neuronal apoptosis and intimal hyperplasia through attenuation of VSMC migration and modulation of matrix metalloproteinase-2/9 and Ras/MEK/ERK signaling pathways. PLoS One 2013; 8:e75654. [PMID: 24086601 PMCID: PMC3782448 DOI: 10.1371/journal.pone.0075654] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 08/16/2013] [Indexed: 12/05/2022] Open
Abstract
Pipoxolan (PIPO) has anti-spasmodic effects, and it is used clinically to relieve smooth muscle spasms. Cerebrovascular disease is one of the leading causes of disability and death worldwide. The main aim of this study was to investigate the effects of PIPO on cerebral ischemia and vascular smooth muscle cell (VSMC) migration in vivo and in vitro. Cerebral infarction area, ratio of intima to media area (I/M ratio) and PCNA antibody staining of the carotid artery in vivo were measured. Cell viability of A7r5 cells, PDGF-BB-stimulated cell migration, and potential mechanisms of PIPO were evaluated by wound healing, transwell and Western blotting. PIPO (10 and 30 mg/kg p.o.) reduced: the cerebral infarction area; neurological deficit; TUNEL-positive cells; cleaved caspase 3-positive cells; intimal hyperplasia; and inhibited proliferating cell nuclear antigen (PCNA)-positive cells in rodents. PIPO (5, 10 and 15 µM) significantly inhibited PDGF-BB-stimulated VSMC migration and reduced Ras, MEK, and p-ERK levels. Moreover, PIPO decreased levels of matrix metalloproteinases -2 and -9 in PDGF-BB-stimulated A7r5 cells. In summary, PIPO is protective in models of ischemia/reperfusion-induced cerebral infarction, carotid artery ligation-induced intimal hyperplasia and VSMC migration both in vivo and in vitro. PIPO could be potentially efficacious in preventing cerebrovascular and vascular diseases.
Collapse
Affiliation(s)
- Yuh-Fung Chen
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan
- * E-mail:
| | - Huei-Yann Tsai
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan
| | - Kuo-Jen Wu
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Lian-Ru Siao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - W. Gibson Wood
- Department of Pharmacology, University of Minnesota and Geriatric Research, Education and Clinical Center, VA Medical Center, Minneapolis, Minnesota, United States of America
| |
Collapse
|
95
|
Wanjare M, Kusuma S, Gerecht S. Perivascular cells in blood vessel regeneration. Biotechnol J 2013; 8:434-47. [PMID: 23554249 DOI: 10.1002/biot.201200199] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/19/2013] [Accepted: 03/05/2013] [Indexed: 12/21/2022]
Abstract
Vascular engineering seeks to design and construct functional blood vessels comprising endothelial cells (ECs) and perivascular cells (PCs), with the ultimate goal of clinical translation. While EC behavior has been extensively investigated, PCs play an equally significant role in the development of novel regenerative strategies, providing functionality and stability to vessels. The two major classes of PCs are vascular smooth muscle cells (vSMCs) and pericytes; vSMCs can be further sub-classified as either contractile or synthetic. The inclusion of these cell types is crucial for successful regeneration of blood vessels. Furthermore, understanding distinctions between vSMCs and pericytes will enable improved therapeutics in a tissue-specific manner. Here we focus on the approaches and challenges facing the use of PCs in vascular regeneration, including their characteristics, stem cell sources, and interactions with ECs. Finally, we discuss biochemical and microRNA (miR) regulators of PC behavior and engineering approaches that mimic various cues affecting PC function.
Collapse
Affiliation(s)
- Maureen Wanjare
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|
96
|
Anti-Proliferative Effect of an Aqueous Extract of Prunella vulgaris in Vascular Smooth Muscle Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:936463. [PMID: 24159354 PMCID: PMC3789443 DOI: 10.1155/2013/936463] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 08/10/2013] [Indexed: 11/18/2022]
Abstract
The abnormal proliferation of vascular smooth muscle cells (VSMCs) in arterial walls is an important pathogenic factor of vascular disorders such as diabetic atherosclerosis. We have reported the anti-inflammatory effect of an aqueous extract from Prunella vulgaris (APV) in vascular endothelial cell. In the present study, APV exhibited inhibitory effects on high glucose-stimulated VSMC proliferation, migration, and invasion activities, inducing G1 cell cycle arrest with downregulation of cyclins and CDKs and upregulation of the CKIs, p21waf1/cip1 and p27kip1. Furthermore, APV dose dependently suppressed the high glucose-induced matrix metalloproteinase activity. High glucose-induced phosphorylation of ERK, p38 MAPK, was decreased by the pretreatment of APV. NF-κB activation by high glucose was attenuated by APV, as an antioxidant. APV attenuated the high glucose-induced decrease of nuclear factor E2-related factor-2 (Nrf2) translocation and heme oxygenase-1 (HO-1) expression. Intracellular cGMP level was also increased by APV treatment. These results demonstrate that APV may inhibit VSMC proliferation via downregulating ROS/NF-κB /ERK/p38 MAPK pathways. In addition, APV has a beneficial effect by the interaction of Nrf2-mediated NO/cGMP with HO-1, suggesting that Prunella vulgaris may be useful in preventing diabetic atherosclerosis.
Collapse
|
97
|
Deatrick KB, Obi A, Luke CE, Elfline MA, Sood V, Upchurch GR, Jaffer F, Wakefield TW, Henke PK. Matrix metalloproteinase-9 deletion is associated with decreased mid-term vein wall fibrosis in experimental stasis DVT. Thromb Res 2013; 132:360-6. [PMID: 23978304 PMCID: PMC3777801 DOI: 10.1016/j.thromres.2013.06.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 06/26/2013] [Accepted: 06/28/2013] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Post thrombotic syndrome therapy is primarily palliative, and the associated vein wall inflammatory mechanisms are unclear. Vein wall fibrotic injury following deep venous thrombosis (VT) is associated with elevated matrix metalloproteinases (MMPs). Whether and by what mechanism MMP9 directly contributes to vein wall remodeling after VT is unknown. METHODS WT and MMP9 -/- mice underwent stasis VT by ligation of the inferior vena cava (IVC) and tissue was harvested at 2, 8, and 21days. Assessment of thrombus size, and gene, protein and structural vein wall determinations were done. RESULTS VT resolution was increased in MMP9-/- mice as compared with controls at 21d only. The primary phenotypic fibrotic vein wall differences occurred at 8d post VT, with significantly less vein wall collagen content as assessed by Picosirius red staining in MMP9 -/- mice as compared with WT. Increased monocytic vein wall influx with less IL-1b and TGFb was found in MMP9 -/- vein walls as compared with WT. Corresponding levels of PAI-1 were increased in MMP9 -/- compared with WT, and no difference in FSP-1+cells as compared with controls. CONCLUSIONS In stasis VT, MMP9 modulates midterm vein wall collagen content, with an altered local inflammatory and profibrotic environment, likely directed by monocytes. Thus, MMP9 plays a role in both vein wall responses as well as late thrombus resolution.
Collapse
Affiliation(s)
- Kristopher B Deatrick
- Conrad Jobst Vascular Research Laboratory, Section of Vascular Surgery, Department of Surgery, University of Michigan Medical School, Boston MA, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Aguilera KY, Brekken RA. Recruitment and retention: factors that affect pericyte migration. Cell Mol Life Sci 2013; 71:299-309. [PMID: 23912898 DOI: 10.1007/s00018-013-1432-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/30/2013] [Accepted: 07/22/2013] [Indexed: 02/07/2023]
Abstract
Pericytes are critical for vascular morphogenesis and contribute to several pathologies, including cancer development and progression. The mechanisms governing pericyte migration and differentiation are complex and have not been fully established. Current literature suggests that platelet-derived growth factor/platelet-derived growth factor receptor-β, sphingosine 1-phosphate/endothelial differentiation gene-1, angiopoietin-1/tyrosine kinase with immunoglobulin-like and EGF-like domains 2, angiopoietin-2/tyrosine kinase with immunoglobulin-like and EGF-like domains 2, transforming growth factor β/activin receptor-like kinase 1, transforming growth factor β/activin receptor-like kinase 5, Semaphorin-3A/Neuropilin, and matrix metalloproteinase activity regulate the recruitment of pericytes to nascent vessels. Interestingly, many of these pathways are directly affected by secreted protein acidic and rich in cysteine (SPARC). Here, we summarize the function of these factors in pericyte migration and discuss if and how SPARC might influence these activities and thus provide an additional layer of control for the recruitment of vascular support cells. Additionally, the consequences of targeted inhibition of pericytes in tumors and the current understanding of pericyte recruitment in pathological environments are discussed.
Collapse
Affiliation(s)
- Kristina Y Aguilera
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75390-8593, USA
| | | |
Collapse
|
99
|
Koenig O, Walker T, Perle N, Zech A, Neumann B, Schlensak C, Wendel HP, Nolte A. New aspects of gene-silencing for the treatment of cardiovascular diseases. Pharmaceuticals (Basel) 2013; 6:881-914. [PMID: 24276320 PMCID: PMC3816708 DOI: 10.3390/ph6070881] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/15/2013] [Accepted: 07/11/2013] [Indexed: 01/17/2023] Open
Abstract
Coronary heart disease (CHD), mainly caused by atherosclerosis, represents the single leading cause of death in industrialized countries. Besides the classical interventional therapies new applications for treatment of vascular wall pathologies are appearing on the horizon. RNA interference (RNAi) represents a novel therapeutic strategy due to sequence-specific gene-silencing through the use of small interfering RNA (siRNA). The modulation of gene expression by short RNAs provides a powerful tool to theoretically silence any disease-related or disease-promoting gene of interest. In this review we outline the RNAi mechanisms, the currently used delivery systems and their possible applications to the cardiovascular system. Especially, the optimization of the targeting and transfection procedures could enhance the efficiency of siRNA delivery drastically and might open the way to clinical applicability. The new findings of the last years may show the techniques to new innovative therapies and could probably play an important role in treating CHD in the future.
Collapse
Affiliation(s)
- Olivia Koenig
- Clinical Research Laboratory, Dept. of Thoracic, Cardiac and Vascular Surgery, University Hospital Tuebingen, Calwerstr. 7/1, 72076 Tuebingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Matrix metalloproteinases: inflammatory regulators of cell behaviors in vascular formation and remodeling. Mediators Inflamm 2013; 2013:928315. [PMID: 23840100 PMCID: PMC3694547 DOI: 10.1155/2013/928315] [Citation(s) in RCA: 279] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
Abnormal angiogenesis and vascular remodeling contribute to pathogenesis of a number of disorders such as tumor, arthritis, atherosclerosis, restenosis, hypertension, and neurodegeneration. During angiogenesis and vascular remodeling, behaviors of stem/progenitor cells, endothelial cells (ECs), and vascular smooth muscle cells (VSMCs) and its interaction with extracellular matrix (ECM) play a critical role in the processes. Matrix metalloproteinases (MMPs), well-known inflammatory mediators are a family of zinc-dependent proteolytic enzymes that degrade various components of ECM and non-ECM molecules mediating tissue remodeling in both physiological and pathological processes. MMPs including MMP-1, MMP-2, MMP-3, MMP-7, MMP-8, MMP-9, MMP-12, and MT1-MMP, are stimulated and activated by various stimuli in vascular tissues. Once activated, MMPs degrade ECM proteins or other related signal molecules to promote recruitment of stem/progenitor cells and facilitate migration and invasion of ECs and VSMCs. Moreover, vascular cell proliferation and apoptosis can also be regulated by MMPs via proteolytically cleaving and modulating bioactive molecules and relevant signaling pathways. Regarding the importance of vascular cells in abnormal angiogenesis and vascular remodeling, regulation of vascular cell behaviors through modulating expression and activation of MMPs shows therapeutic potential.
Collapse
|