51
|
Su M, Yue Z, Wang H, Jia M, Bai C, Qiu W, Chen J. Ufmylation Is Activated in Vascular Remodeling and Lipopolysaccharide-Induced Endothelial Cell Injury. DNA Cell Biol 2018; 37:426-431. [PMID: 29461087 DOI: 10.1089/dna.2017.4073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ming Su
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Zhihong Yue
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Mei Jia
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, People's Republic of China
| | - Congxia Bai
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Wei Qiu
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jingzhou Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
52
|
Wang D, Uhrin P, Mocan A, Waltenberger B, Breuss JM, Tewari D, Mihaly-Bison J, Huminiecki Ł, Starzyński RR, Tzvetkov NT, Horbańczuk J, Atanasov AG. Vascular smooth muscle cell proliferation as a therapeutic target. Part 1: molecular targets and pathways. Biotechnol Adv 2018; 36:1586-1607. [PMID: 29684502 DOI: 10.1016/j.biotechadv.2018.04.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases are a major cause of human death worldwide. Excessive proliferation of vascular smooth muscle cells contributes to the etiology of such diseases, including atherosclerosis, restenosis, and pulmonary hypertension. The control of vascular cell proliferation is complex and encompasses interactions of many regulatory molecules and signaling pathways. Herein, we recapitulated the importance of signaling cascades relevant for the regulation of vascular cell proliferation. Detailed understanding of the mechanism underlying this process is essential for the identification of new lead compounds (e.g., natural products) for vascular therapies.
Collapse
Affiliation(s)
- Dongdong Wang
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria; Institute of Clinical Chemistry, University Hospital Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Hațieganu" University of Medicine and Pharmacy, Strada Gheorghe Marinescu 23, 400337 Cluj-Napoca, Romania; Institute for Life Sciences, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Johannes M Breuss
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Devesh Tewari
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Bhimtal, 263136 Nainital, Uttarakhand, India
| | - Judit Mihaly-Bison
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Łukasz Huminiecki
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Rafał R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; NTZ Lab Ltd., Krasno Selo 198, 1618 Sofia, Bulgaria
| | - Jarosław Horbańczuk
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| |
Collapse
|
53
|
Jung TW, Park HS, Choi GH, Kim D, Lee T. CTRP9 Regulates Growth, Differentiation, and Apoptosis in Human Keratinocytes through TGFβ1-p38-Dependent Pathway. Mol Cells 2017; 40:906-915. [PMID: 29145717 PMCID: PMC5750709 DOI: 10.14348/molcells.2017.0097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 11/29/2022] Open
Abstract
Impairment of wound healing is a common problem in individuals with diabetes. Adiponectin, an adipocyte-derived cytokine, has many beneficial effects on metabolic disorders such as diabetes, obesity, hypertension, and dyslipidemia. C1q/TNF-Related Protein 9 (CTRP9), the closest paralog of adiponectin, has been reported to have beneficial effects on wound healing. In the current study, we demonstrate that CTRP9 regulates growth, differentiation, and apoptosis of HaCaT human keratinocytes. We found that CTRP9 augmented expression of transforming growth factor beta 1 (TGFβ1) by transcription factor activator protein 1 (AP-1) binding activity and phosphorylation of p38 in a dose-dependent manner. Furthermore, siRNA-mediated suppression of TGFβ1 reversed the increase in p38 phosphorylation induced by CTRP9. siRNA-mediated suppression of TGFβ1 or p38 significantly abrogated the effects of CTRP9 on cell proliferation and differentiation while inducing apoptosis, implying that CTRP9 stimulates wound recovery through a TGFβ1-dependent pathway in keratinocytes. Furthermore, intravenous injection of CTRP9 via tail vein suppressed mRNA expression of Ki67 and involucrin whereas it augmented TGFβ1 mRNA expression and caspase 3 activity in skin of type 1 diabetes animal models. In conclusion, our results suggest that CTRP9 has suppressive effects on hyperkeratosis, providing a potentially effective therapeutic strategy for diabetic wounds.
Collapse
Affiliation(s)
- Tae Woo Jung
- Research Administration Team, Seoul National University Bundang Hospital, Seongnam 13620,
Korea
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620,
Korea
| | - Hyung Sub Park
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620,
Korea
| | - Geum Hee Choi
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620,
Korea
| | - Daehwan Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620,
Korea
| | - Taeseung Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620,
Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080,
Korea
| |
Collapse
|
54
|
Kim HS, Kim SK, Kang KW. Differential Effects of sEH Inhibitors on the Proliferation and Migration of Vascular Smooth Muscle Cells. Int J Mol Sci 2017; 18:ijms18122683. [PMID: 29232926 PMCID: PMC5751285 DOI: 10.3390/ijms18122683] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/30/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023] Open
Abstract
Epoxyeicosatrienoic acid (EET) is a cardioprotective metabolite of arachidonic acid. It is known that soluble epoxide hydrolase (sEH) is involved in the metabolic degradation of EET. The abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) play important roles in the pathogenesis of atherosclerosis and restenosis. Thus, the present study investigated the effects of the sEH inhibitor 12-(((tricyclo(3.3.1.13,7)dec-1-ylamino)carbonyl)amino)-dodecanoic acid (AUDA) on platelet-derived growth factor (PDGF)-induced proliferation and migration in rat VSMCs. AUDA significantly inhibited PDGF-induced rat VSMC proliferation, which coincided with Pin1 suppression and heme oxygenase-1 (HO-1) upregulation. However, exogenous 8,9-EET, 11,12-EET, and 14,15-EET treatments did not alter Pin1 or HO-1 levels and had little effect on the proliferation of rat VSMCs. On the other hand, AUDA enhanced the PDGF-stimulated cell migration of rat VSMCs. Furthermore, AUDA-induced activation of cyclooxygenase-2 (COX-2) and subsequent thromboxane A2 (TXA2) production were required for the enhanced migration. Additionally, EETs increased COX-2 expression but inhibited the migration of rat VSMCs. In conclusion, the present study showed that AUDA exerted differential effects on the proliferation and migration of PDGF-stimulated rat VSMCs and that these results may not depend on EET stabilization.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Enzyme Inhibitors/pharmacology
- Epoxide Hydrolases/antagonists & inhibitors
- Epoxy Compounds/metabolism
- Gene Expression Regulation/drug effects
- Heme Oxygenase-1/metabolism
- Lauric Acids/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Rats
Collapse
Affiliation(s)
- Hyo Seon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea.
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
55
|
Chang Y, Li JY, Jayakumar T, Hung SH, Lee WC, Manubolu M, Sheu JR, Hsu MJ. Ketamine, a Clinically Used Anesthetic, Inhibits Vascular Smooth Muscle Cell Proliferation via PP2A-Activated PI3K/Akt/ERK Inhibition. Int J Mol Sci 2017; 18:ijms18122545. [PMID: 29186909 PMCID: PMC5751148 DOI: 10.3390/ijms18122545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 01/09/2023] Open
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) gives rise to major pathological processes involved in the development of cardiovascular diseases. The use of anti-proliferative agents for VSMCs offers potential for the treatment of vascular disorders. Intravenous anesthetics are firmly established to have direct effects on VSMCs, resulting in modulation of blood pressure. Ketamine has been used for many years in the intensive care unit (ICU) for sedation, and has recently been considered for adjunctive therapy. In the present study, we investigated the effects of ketamine on platelet-derived growth factor BB (PDGF-BB)-induced VSMC proliferation and the associated mechanism. Ketamine concentration-dependently inhibited PDGF-BB-induced VSMC proliferation without cytotoxicity, and phosphatidylinositol 3-kinase (PI3K) and extracellular signal-regulated protein kinase (ERK) inhibitors, LY294002 and PD98059, respectively, have similar inhibitory effects. Ketamine was shown to attenuate PI3K, Akt, and ERK1/2 phosphorylation induced by PDGF-BB. Okadaic acid, a selective protein phosphatase 2A (PP2A) inhibitor, significantly reversed ketamine-mediated PDGF-BB-induced PI3K, Akt, and ERK1/2 phosphorylation; a transfected protein phosphatse 2a (pp2a) siRNA reversed Akt and ERK1/2 phosphorylation; and 3-O-Methyl-sphingomyeline (3-OME), an inhibitor of sphingomyelinase, also significantly reversed ERK1/2 phosphorylation. Moreover, ketamine alone significantly inhibited tyrosine phosphorylation and demethylation of PP2A in a concentration-dependent manner. In addition, the pp2a siRNA potently reversed the ketamine-activated catalytic subunit (PP2A-C) of PP2A. These results provide evidence of an anti-proliferating effect of ketamine in VSMCs, showing activation of PP2A blocks PI3K, Akt, and ERK phosphorylation that subsequently inhibits the proliferation of VSMCs. Thus, ketamine may be considered a potential effective therapeutic agent for reducing atherosclerotic process by blocking the proliferation of VSMCs.
Collapse
Affiliation(s)
- Yi Chang
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, No. 95, Wenchang Rd., Taipei 111, Taiwan.
- School of Medicine, Fu-Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City 242, Taiwan.
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu-Hsing St., Taipei 110, Taiwan.
| | - Jiun-Yi Li
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu-Hsing St., Taipei 110, Taiwan.
- Department of Cardiovascular Surgery, Mackay Memorial Hospital, and Mackay Medical College, Taipei 104, Taiwan.
| | - Thanasekaran Jayakumar
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu-Hsing St., Taipei 110, Taiwan.
| | - Shou-Huang Hung
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu-Hsing St., Taipei 110, Taiwan.
| | - Wei-Cheng Lee
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu-Hsing St., Taipei 110, Taiwan.
| | - Manjunath Manubolu
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, 1314 Kinnear Rd, Columbus, OH 43212, USA.
| | - Joen-Rong Sheu
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu-Hsing St., Taipei 110, Taiwan.
| | - Ming-Jen Hsu
- Department of Pharmacology and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu-Hsing St., Taipei 110, Taiwan.
| |
Collapse
|
56
|
Zhang Y, Cui Y, Deng W, Wang H, Qin W, Huang C, Li C, Zhang J, Guo Y, Wu D, Guo H. Isoquercitrin protects against pulmonary hypertension via inhibiting PASMCs proliferation. Clin Exp Pharmacol Physiol 2017; 44:362-370. [PMID: 27873355 DOI: 10.1111/1440-1681.12705] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/06/2016] [Accepted: 11/18/2016] [Indexed: 12/21/2022]
Abstract
Pulmonary vascular remodelling is a common feature among the heterogeneous disorders that cause pulmonary arterial hypertension (PAH), and pulmonary arterial smooth muscle cells (PASMCs) proliferation impact the long-term prognosis of the patient. Isoquercitrin (IQC) is a flavonoid with anti-oxidative, anti-inflammatory and anti-proliferative activations. This study aimed to investigate whether IQC could prevent PASMCs proliferation and vascular remodelling in monocrotaline (MCT) induced PAH. Male Wistar rats were administered with Vehicle or 0.1% IQC maintain feed after MCT (40 mg/kg) injection. Haemodynamic changes, right ventricular hypertrophy and lung morphological features were assessed 3 weeks later. MCT-induced PAH, pulmonary vascular remodelling and PASMCs proliferation in Vehicle-treated rats. IQC reduced the right ventricle systolic pressure (RVSP), the ratio of RV/LV+S and the RV hypertrophy. IQC significantly alleviated the expression of proliferating cell nuclear antigen (PCNA), smooth muscle α-actin (α-SMA), and the percentage of fully muscularized small arterioles. In vitro studies, PASMCs were pretreated with IQC and stimulated with platelet-derived growth factor (PDGF)-BB (20 ng/mL). IQC suppressed PDGF-BB-induced PASMCs proliferation and caused G0/G1 phase cell cycle arrest. IQC downregulated the expression of Cyclin D1 and CDK4 as well as inhibited p27Kip1 degradation. Meanwhile, IQC negatively modulated PDGF-BB-induced phosphorylation of PDGF-Rβ, Akt/GSK3β and ERK1/2. IQC ameliorated MCT-induced pulmonary vascular remodelling via suppressing PASMCs proliferation and blocking PDGF-Rβ signalling pathway.
Collapse
Affiliation(s)
- Yongtao Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuqian Cui
- Centre for Reproductive Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Weidong Qin
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Chengmin Huang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Li
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Jianning Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yuan Guo
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Dawei Wu
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Haipeng Guo
- Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
57
|
Song IS, Jeong YJ, Park JH, Shim S, Jang SW. Chebulinic acid inhibits smooth muscle cell migration by suppressing PDGF-Rβ phosphorylation and inhibiting matrix metalloproteinase-2 expression. Sci Rep 2017; 7:11797. [PMID: 28924208 PMCID: PMC5603554 DOI: 10.1038/s41598-017-12221-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/06/2017] [Indexed: 01/10/2023] Open
Abstract
Excessive migration of vascular smooth muscle cells (VSMCs) after vascular injury contributes to the development of occlusive vascular disease. Inhibition of VSMC migration is a validated therapeutic modality for occlusive vascular diseases, such as atherosclerosis and restenosis. We investigated the inhibitory effect of chebulinic acid (CBA) on cell migration and matrix metalloproteinase (MMP)-2 activation in platelet-derived growth factor (PDGF)-BB-induced mouse and human VSMCs. CBA significantly inhibited PDGF-BB-induced migration in mouse and human VSMCs, without inducing cell death. Additionally, CBA significantly blocked PDGF-BB-induced phosphorylation of the PDGF receptor (PDGF-R), Akt, and extracellular signal-regulated kinase (ERK)1/2 by inhibiting the activation of the PDGF-BB signalling pathway. In both mouse and human VSMCs, CBA inhibited PDGF-induced MMP-2 mRNA and protein expression as well as the proteolytic activity of MMP-2. Moreover, CBA suppressed sprout outgrowth formation of VSMCs from endothelium-removed aortic rings as well as neointima formation following rat carotid balloon injury. Taken together, our findings indicated that CBA inhibits VSMC migration by decreasing MMP-2 expression through PDGF-R and the ERK1/2 and Akt pathways. Our data may improve the understanding of the antiatherogenic effects of CBA in VSMCs.
Collapse
Affiliation(s)
- In-Sung Song
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea.,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | - Yu Jeong Jeong
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea.,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | - Jung-Hyun Park
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea.,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | - Sungbo Shim
- Department of Biochemistry, Neuromarker Resource Bank, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea.
| | - Sung-Wuk Jang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 138-736, Korea. .,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 138-736, Korea.
| |
Collapse
|
58
|
Greig FH, Kennedy S, Gibson G, Ramos JW, Nixon GF. PEA-15 (Phosphoprotein Enriched in Astrocytes 15) Is a Protective Mediator in the Vasculature and Is Regulated During Neointimal Hyperplasia. J Am Heart Assoc 2017; 6:JAHA.117.006936. [PMID: 28893763 PMCID: PMC5634313 DOI: 10.1161/jaha.117.006936] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Neointimal hyperplasia following angioplasty occurs via vascular smooth muscle cell proliferation. The mechanisms involved are not fully understood but include mitogen-activated protein kinases ERK1/2 (extracellular signal-regulated kinases 1 and 2). We recently identified the intracellular mediator PEA-15 (phosphoprotein enriched in astrocytes 15) in vascular smooth muscle cells as a regulator of ERK1/2-dependent proliferation in vitro. PEA-15 acts as a cytoplasmic anchor for ERK1/2, preventing nuclear localization and thereby reducing ERK1/2-dependent gene expression. The aim of the current study was to examine the role of PEA-15 in neointimal hyperplasia in vivo. METHOD AND RESULTS Mice deficient in PEA-15 or wild-type mice were subjected to wire injury of the carotid artery. In uninjured arteries from PEA-15-deficient mice, ERK1/2 had increased nuclear translocation and increased basal ERK1/2-dependent transcription. Following wire injury, arteries from PEA-15-deficient mice developed neointimal hyperplasia at an increased rate compared with wild-type mice. This occurred in parallel with an increase in a proliferative marker and vascular smooth muscle cell proliferation. In wild-type mice, PEA-15 expression was decreased in vascular smooth muscle cells at an early stage before any increase in intima:media ratio. This regulation of PEA-15 expression following injury was also observed in an ex vivo human model of hyperplasia. CONCLUSIONS These results indicate, for the first time, a novel protective role for PEA-15 against inappropriate vascular proliferation. PEA-15 expression may also be repressed during vascular injury, suggesting that maintenance of PEA-15 expression is a novel therapeutic target in vascular disease.
Collapse
Affiliation(s)
- Fiona H Greig
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, United Kingdom
| | - Simon Kennedy
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - George Gibson
- Department of Cardiothoracic Surgery, Aberdeen Royal Hospital, Aberdeen, United Kingdom
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Centre University of Hawaii at Mānoa, Honolulu, HI
| | - Graeme F Nixon
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, United Kingdom
| |
Collapse
|
59
|
Lietman CD, Segedy AK, Li B, Fazio S, Atkinson JB, Linton MF, Young PP. Loss of SPRR3 in ApoE-/- mice leads to atheroma vulnerability through Akt dependent and independent effects in VSMCs. PLoS One 2017; 12:e0184620. [PMID: 28886156 PMCID: PMC5590986 DOI: 10.1371/journal.pone.0184620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/28/2017] [Indexed: 01/18/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) represent important modulators of plaque stability in advanced lesions. We previously reported that loss of small proline-rich repeat protein 3 (Sprr3), leads to VSMC apoptosis in a PI3K/Akt-dependent manner and accelerates lesion progression. Here, we investigated the role of Sprr3 in modulating plaque stability in hyperlipidemic ApoE-/- mice. We show that loss of Sprr3 increased necrotic core size and reduced cap collagen content of atheromas in brachiocephalic arteries with evidence of plaque rupture and development of intraluminal thrombi. Moreover, Sprr3-/-ApoE-/- mice developed advanced coronary artery lesions accompanied by intraplaque hemorrhage and left ventricle microinfarcts. SPRR3 is known to reduce VSMC survival in lesions by promoting their apoptosis. In addition, we demonstrated that Sprr3-/- VSMCs displayed reduced expression of procollagen in a PI3K/Akt dependent manner. SPRR3 loss also increased MMP gelatinase activity in lesions, and increased MMP2 expression, migration and contraction of VSMCs independently of PI3K/Akt. Consequently, Sprr3 represents the first described VSMC modulator of each of the critical features of cap stability, including VSMC numbers, collagen type I synthesis, and protease activity through Akt dependent and independent pathways.
Collapse
Affiliation(s)
- Caressa D. Lietman
- Department of Pathology Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, United States of America
| | - Amanda K. Segedy
- Department of Pathology Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, United States of America
| | - Bin Li
- Department of Pathology Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, United States of America
| | - Sergio Fazio
- Center of Preventive Cardiology; Knight Cardiovascular Institute; Oregon Health & Science University; Portland, OR, United States of America
| | - James B. Atkinson
- Department of Pathology Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, United States of America
- Veterans Affairs Medical Center, Nashville, TN, United States of America
| | - MacRae F. Linton
- Department of Pharmacology, Vanderbilt University Medical Center; Nashville, TN, United States of America
- Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, United States of America
| | - Pampee P. Young
- Department of Pathology Microbiology and Immunology; Vanderbilt University Medical Center; Nashville, TN, United States of America
- Veterans Affairs Medical Center, Nashville, TN, United States of America
- Department of Medicine; Vanderbilt University Medical Center; Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
60
|
Zhou P, Lu S, Luo Y, Wang S, Yang K, Zhai Y, Sun G, Sun X. Attenuation of TNF-α-Induced Inflammatory Injury in Endothelial Cells by Ginsenoside Rb1 via Inhibiting NF-κB, JNK and p38 Signaling Pathways. Front Pharmacol 2017; 8:464. [PMID: 28824425 PMCID: PMC5540891 DOI: 10.3389/fphar.2017.00464] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/30/2017] [Indexed: 12/18/2022] Open
Abstract
It is currently believed that inflammation plays a central role in the pathophysiology of atherosclerosis. Oxidative stress and redox-sensitive transcription factors are implicated in the process. Ginsenoside Rb1, a major active ingredient in processed Radix notoginseng, has attracted widespread attention because of its potential to improve cardiovascular function. However, the effects of ginsenoside Rb1 on tumor necrosis factor-α (TNF-α)-induced vascular endothelial cell injury and the underlying molecular mechanisms have never been studied. This study showed that TNF-α-induced oxidative stress, inflammation and apoptosis in human umbilical vein endothelial cells (HUVECs) could be attenuated by ginsenoside Rb1 pretreatment. Using JC-1, Annexin V/PI and TUNEL staining, and a caspase-3 activity assay, we found that Rb1 provided significant protection against TNF-α-induced cell death. Furthermore, Rb1 pretreatment could inhibit TNF-α-induced ROS and MDA production; increase the activities of SOD, CAT, and GSH-Px; and decrease the levels of IL-1β, IL-6, VCAM-1, ICAM-1, VEGF, MMP-2 and MMP-9. Importantly, the cytoprotective effects of Rb1 were correlated with NF-κB signaling pathway inhibition. Additionally, we found that Rb1 may suppress the NF-κB pathway through p-38 and JNK pathway activation, findings supported by the results of our experiments involving anisomycin (AM), a JNK and p38 activator. In conclusion, this study showed that ginsenoside Rb1 protects HUVECs from TNF-α-induced oxidative stress and inflammation by inhibiting JNK and p38. This inhibition suppressed NF-κB signaling and down-regulated the expression of inflammatory factors and apoptosis-related proteins.
Collapse
Affiliation(s)
- Ping Zhou
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| | - Shan Lu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| | - Shan Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| | - Ke Yang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| | - Yadong Zhai
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical SciencesBeijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineBeijing, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of EducationBeijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese MedicineBeijing, China
| |
Collapse
|
61
|
Wu W, Zhang J, Zhao C, Sun Y, Pang W, Yang G. CTRP6 Regulates Porcine Adipocyte Proliferation and Differentiation by the AdipoR1/MAPK Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:5512-5522. [PMID: 28535682 DOI: 10.1021/acs.jafc.7b00594] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Intramuscular fat (IMF) and subcutaneous fat (SCF), which are modulated by adipogenesis of intramuscular and subcutaneous adipocytes, play key roles in pork quality. C1q/tumor necrosis factor-related protein 6 (CTRP6), an adipokine, plays an important role in the differentiation of 3T3-L1 cells. However, the effect and regulatory mechanisms of CTRP6 on porcine adipogenesis, and whether CTRP6 has the same effect on intramuscular and subcutaneous adipocytes, are still unknown. Here, we found that CTRP6 significantly inhibited both adipocyte proliferation assessed by proliferative marker expression, but CTRP6 decreased the proliferation rate of intramuscular adipocytes (IM) to a greater extent than subcutaneous adipocytes (SC). Moreover, CTRP6 promoted the activity of the p38 signaling pathway during the proliferation of both cell types. Nevertheless, in subcutaneous adipocytes, CTRP6 also influenced the phosphorylation of extracellular regulated protein kinases1/2 (p-Erk1/2), but not in intramuscular adipocytes. Additionally, during the differentiation of intramuscular and subcutaneous adipocytes, CTRP6 increased adipogenic genes expression and the level of p-p38, while it decreased the activity of p-Erk1/2. Interestingly, the effect of CTRP6 shRNA or CTRP6 recombinant protein was attenuated by U0126 (a special p-Erk inhibitor) or SB203580 (a special p-p38 inhibitor) in adipocytes. By target gene prediction and experimental validation, we demonstrated that CTRP6 may be a target of miR-29a in porcine adipocytes. Moreover, AdipoR1was identified as a receptor of CTRP6 in intramuscular adipocytes, but not in subcutaneous adipocytes. On the basis of the above findings, we suggest that CTRP6 was the target gene of miR-29a, inhibited intramuscular and subcutaneous adipocyte proliferation, but promoted differentiation by the mitogen-activated protein kinase (MAPK) signaling pathway. These findings indicate that CTRP6 played an essentially regulatory role in fat development.
Collapse
Affiliation(s)
- Wenjing Wu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University , Yangling, Shaanxi 712100, China
- College of Biological and Chemical Engineering, Jiaxing University , Jiaxing, Zhejiang 314000, China
| | - Jin Zhang
- College of Biological and Chemical Engineering, Jiaxing University , Jiaxing, Zhejiang 314000, China
| | - Chen Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University , Yangling, Shaanxi 712100, China
| | - Yunmei Sun
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University , Yangling, Shaanxi 712100, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University , Yangling, Shaanxi 712100, China
| | - Gongshe Yang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University , Yangling, Shaanxi 712100, China
| |
Collapse
|
62
|
Abou Ziki MD, Mani A. Wnt signaling, a novel pathway regulating blood pressure? State of the art review. Atherosclerosis 2017; 262:171-178. [PMID: 28522145 PMCID: PMC5508596 DOI: 10.1016/j.atherosclerosis.2017.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/06/2017] [Accepted: 05/03/2017] [Indexed: 12/18/2022]
Abstract
Recent antihypertensive trials show conflicting results on blood pressure (BP) targets in patient populations with different metabolic profiles, with lowest benefit from tight BP control observed in patients with type 2 diabetes mellitus. This paradox could arise from the heterogeneity of study populations and underscores the importance of precision medicine initiatives towards understanding and treating hypertension. Wnt signaling pathways and genetic variations in its signaling peptides have been recently associated with metabolic syndrome, hypertension and diabetes, generating a breakthrough for advancement of precision medicine in the field of hypertension. We performed a review of PubMed for publications addressing the contributions of Wnt to BP regulation and hypertension. In addition, we performed a manual search of the reference lists for relevant articles, and included unpublished observations from our laboratory. There is emerging evidence for Wnt's role in BP regulation and its involvement in the pathogenesis of hypertension. Wnt signaling has pleiotropic effects on distinct pathways that involve vascular smooth muscle plasticity, and cardiac, renal, and neural physiology. Hypertension is a heterogeneous disease with unique molecular pathways regulating its response to therapy. Recognition of these pathways is a prerequisite to identify novel targets for drug development and personalizing medicine. A review of Wnt signaling reveals its emerging role in BP regulation and as a target for novel drug development that has the potential to transform the therapy of hypertension in specific populations.
Collapse
Affiliation(s)
- Maen D Abou Ziki
- Departments of Internal Medicine and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Arya Mani
- Departments of Internal Medicine and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
63
|
Yao Y, Hu Z, Ye J, Hu C, Song Q, Da X, Yu Y, Li H, Xu C, Chen Q, Wang QK. Targeting AGGF1 (angiogenic factor with G patch and FHA domains 1) for Blocking Neointimal Formation After Vascular Injury. J Am Heart Assoc 2017. [PMID: 28649088 PMCID: PMC5669188 DOI: 10.1161/jaha.117.005889] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Despite recent improvements in angioplasty and placement of drug‐eluting stents in treatment of atherosclerosis, restenosis and in‐stent thrombosis impede treatment efficacy and cause numerous deaths. Research efforts are needed to identify new molecular targets for blocking restenosis. We aim to establish angiogenic factor AGGF1 (angiogenic factor with G patch and FHA domains 1) as a novel target for blocking neointimal formation and restenosis after vascular injury. Methods and Results AGGF1 shows strong expression in carotid arteries; however, its expression is markedly decreased in arteries after vascular injury. AGGF1+/− mice show increased neointimal formation accompanied with increased proliferation of vascular smooth muscle cells (VSMCs) in carotid arteries after vascular injury. Importantly, AGGF1 protein therapy blocks neointimal formation after vascular injury by inhibiting the proliferation and promoting phenotypic switching of VSMCs to the contractile phenotype in mice in vivo. In vitro, AGGF1 significantly inhibits VSMCs proliferation and decreases the cell numbers at the S phase. AGGF1 also blocks platelet‐derived growth factor‐BB–induced proliferation, migration of VSMCs, increases expression of cyclin D, and decreases expression of p21 and p27. AGGF1 inhibits phenotypic switching of VSMCs to the synthetic phenotype by countering the inhibitory effect of platelet‐derived growth factor‐BB on SRF expression and the formation of the myocardin/SRF/CArG‐box complex involved in activation of VSMCs markers. Finally, we show that AGGF1 inhibits platelet‐derived growth factor‐BB–induced phosphorylation of MEK1/2, ERK1/2, and Elk phosphorylation involved in the phenotypic switching of VSMCs, and that overexpression of Elk abolishes the effect of AGGF1. Conclusions AGGF1 protein therapy is effective in blocking neointimal formation after vascular injury by regulating a novel AGGF1‐MEK1/2‐ERK1/2‐Elk‐myocardin‐SRF/p27 signaling pathway.
Collapse
Affiliation(s)
- Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenkun Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Ye
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Changqing Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qixue Song
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Xingwen Da
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Yubin Yu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyun Chen
- Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH
| | - Qing Kenneth Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China .,Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
64
|
Quantitative Phosphoproteomics Reveals a Role for Collapsin Response Mediator Protein 2 in PDGF-Induced Cell Migration. Sci Rep 2017. [PMID: 28638064 PMCID: PMC5479788 DOI: 10.1038/s41598-017-04015-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The Platelet Derived Growth Factor (PDGF) family of ligands have well established functions in the induction of cell proliferation and migration during development, tissue homeostasis and interactions between tumours and stroma. However, the mechanisms by which these actions are executed are incompletely understood. Here we report a differential phosphoproteomics study, using a SILAC approach, of PDGF-stimulated mouse embryonic fibroblasts (MEFs). 116 phospho-sites were identified as up-regulated and 45 down-regulated in response to PDGF stimulation. These encompass proteins involved in cell adhesion, cytoskeleton regulation and vesicle-mediated transport, significantly expanding the range of proteins implicated in PDGF signalling pathways. Included in the down-regulated class was the microtubule bundling protein Collapsin Response Mediator Protein 2 (CRMP2). In response to stimulation with PDGF, CRMP2 was dephosphorylated on Thr514, an event known to increase CRMP2 activity. This was reversed in the presence of micromolar concentrations of the protein phosphatase inhibitor okadaic acid, implicating PDGF-induced activation of protein phosphatase 1 (PP1) in CRMP2 regulation. Depletion of CRMP2 resulted in impairment of PDGF-mediated cell migration in an in vitro wound healing assay. These results show that CRMP2 is required for PDGF-directed cell migration in vitro.
Collapse
|
65
|
Heggermont WA, Papageorgiou AP, Quaegebeur A, Deckx S, Carai P, Verhesen W, Eelen G, Schoors S, van Leeuwen R, Alekseev S, Elzenaar I, Vinckier S, Pokreisz P, Walravens AS, Gijsbers R, Van Den Haute C, Nickel A, Schroen B, van Bilsen M, Janssens S, Maack C, Pinto Y, Carmeliet P, Heymans S. Inhibition of MicroRNA-146a and Overexpression of Its Target Dihydrolipoyl Succinyltransferase Protect Against Pressure Overload-Induced Cardiac Hypertrophy and Dysfunction. Circulation 2017; 136:747-761. [PMID: 28611091 DOI: 10.1161/circulationaha.116.024171] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 05/10/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiovascular diseases remain the predominant cause of death worldwide, with the prevalence of heart failure continuing to increase. Despite increased knowledge of the metabolic alterations that occur in heart failure, novel therapies to treat the observed metabolic disturbances are still lacking. METHODS Mice were subjected to pressure overload by means of angiotensin-II infusion or transversal aortic constriction. MicroRNA-146a was either genetically or pharmacologically knocked out or genetically overexpressed in cardiomyocytes. Furthermore, overexpression of dihydrolipoyl succinyltransferase (DLST) in the murine heart was performed by means of an adeno-associated virus. RESULTS MicroRNA-146a was upregulated in whole heart tissue in multiple murine pressure overload models. Also, microRNA-146a levels were moderately increased in left ventricular biopsies of patients with aortic stenosis. Overexpression of microRNA-146a in cardiomyocytes provoked cardiac hypertrophy and left ventricular dysfunction in vivo, whereas genetic knockdown or pharmacological blockade of microRNA-146a blunted the hypertrophic response and attenuated cardiac dysfunction in vivo. Mechanistically, microRNA-146a reduced its target DLST-the E2 subcomponent of the α-ketoglutarate dehydrogenase complex, a rate-controlling tricarboxylic acid cycle enzyme. DLST protein levels significantly decreased on pressure overload in wild-type mice, paralleling a decreased oxidative metabolism, whereas DLST protein levels and hence oxidative metabolism were partially maintained in microRNA-146a knockout mice. Moreover, overexpression of DLST in wild-type mice protected against cardiac hypertrophy and dysfunction in vivo. CONCLUSIONS Altogether we show that the microRNA-146a and its target DLST are important metabolic players in left ventricular dysfunction.
Collapse
Affiliation(s)
- Ward A Heggermont
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Anna-Pia Papageorgiou
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Annelies Quaegebeur
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Sophie Deckx
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Paolo Carai
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Wouter Verhesen
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Guy Eelen
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Sandra Schoors
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Rick van Leeuwen
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Sergey Alekseev
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Ies Elzenaar
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Stefan Vinckier
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Peter Pokreisz
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Ann-Sophie Walravens
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Rik Gijsbers
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Chris Van Den Haute
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Alexander Nickel
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Blanche Schroen
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Marc van Bilsen
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Stefan Janssens
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Christoph Maack
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Yigal Pinto
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Peter Carmeliet
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.)
| | - Stephane Heymans
- From Center for Molecular and Vascular Research, Leuven, Belgium (W.H., A.P., S.D., Pa.C., P.P., A.S.W., S.J., S.H.); Center for Heart Failure Research, Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, The Netherlands (W.H., A.P., S.D., Pa.C., W.V., R.v.L., B.S., M.v.B., S.H.); Cardiovascular Research Center, OLV Hospital, Aalst, Belgium (W.H.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium (A.Q., G.E., S.S., S.V., Pe.C.); Amsterdam Medical Center, Amsterdam University, The Netherlands (S.A., I.E., Y.P.); Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences (R.G., C.V.D.H.), Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences (R.G., C.V.D.H.), Leuven Viral Vector Core, Belgium (R.G., C.V.D.H.); and Klinik für Innere Medezin III, Universitätsklinikum des Saarlandes, Homburg, Germany (A.N., C.M.).
| |
Collapse
|
66
|
Ouyang L, Zhang K, Chen J, Wang J, Huang H. Roles of platelet-derived growth factor in vascular calcification. J Cell Physiol 2017; 233:2804-2814. [PMID: 28467642 DOI: 10.1002/jcp.25985] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is prevalent in aging, and patients with hypertension, chronic kidney disease (CKD), or diabetes. VC is regarded as an active and complex process that involves multiple mechanisms responsible for calcium deposition in vessel wall. In light of the complicated pathogenesis of VC, effective therapy for ameliorating VC is limited. Thus, it is urgent to explore the potential mechanisms and find new targets for the therapy of VC. Platelet-derived growth factor (PDGF), a potent mitogen, and chemoattractant have been found to disturb the vascular homeostasis by inducing inflammation, oxidative stress, and phenotype transition, all of which accelerate the process of VC. The aim of current review is to present a review about the roles of PDGF in affecting VC and to establish a potential target for treating VC.
Collapse
Affiliation(s)
- Liu Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Kun Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Jie Chen
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingfeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| | - Hui Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong Province, China
| |
Collapse
|
67
|
de Oliveira MG, Doro FG, Tfouni E, Krieger MH. Phenotypic switching prevention and proliferation/migration inhibition of vascular smooth muscle cells by the ruthenium nitrosyl complex trans-[Ru(NO)Cl(cyclam](PF 6 ) 2. ACTA ACUST UNITED AC 2017; 69:1155-1165. [PMID: 28590566 DOI: 10.1111/jphp.12755] [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] [Received: 12/04/2016] [Accepted: 05/07/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Vascular smooth muscle cell (VSMC) migration and proliferation at sites of vascular injury are both critical steps in the development of intimal hyperplasia (IH). Local delivery of nitric oxide (NO) largely prevents these events. Among the NO donors, tetraazamacrocyclic nitrosyl complexes, such as trans-[Ru(NO)Cl(cyclam)](PF6 )2 (cyclamNO), gained attention for their features, which include the possibility of being embedded in solid matrices, and ability to participate in a nitrite/NO catalytic conversion cycle. METHODS Methods used to evaluate cyclamNO activity: safety margin by NR and MTT; cell proliferation by 3H-thymidine incorporation and proliferating cell nuclear antigen (PCNA) expression; antimigratory properties by transwell and wound healing; prevention of cell phenotypic switching under platelet-derived growth factor type BB (PDGF-BB) stimuli by analysis of alpha smooth muscle actin (α-SMA) expression. KEY FINDINGS Cell proliferation and migration induced by PDGF-BB were significantly inhibited by cyclamNO. The ~60% reduction on expression of contractile protein α-SMA induced by PDGF-BB revealed VSMC phenotypic switching which is significantly prevented by cyclamNO. Compared to the NO donor sodium nitroprusside, cyclamNO showed to be significantly less cytotoxic. CONCLUSIONS With great potential to maintain VSMC functionality and prevent IH-associated events, cyclamNO might be a promissory drug for several applications in cardiovascular medicine, as in stents.
Collapse
Affiliation(s)
- Mariana G de Oliveira
- Laboratório de Cardiovascular, Departamento de Anatomia, Biologia Celular e Fisiologia, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Fabio G Doro
- Departamento de Química Geral e Inorgânica, Instituto de Química, Universidade Federal da Bahia (UFBA), Salvador, BA, Brazil
| | - Elia Tfouni
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Marta H Krieger
- Laboratório de Cardiovascular, Departamento de Anatomia, Biologia Celular e Fisiologia, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
68
|
Wang YC, Cui XB, Chuang YH, Chen SY. Janus Kinase 3, a Novel Regulator for Smooth Muscle Proliferation and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2017; 37:1352-1360. [PMID: 28473442 DOI: 10.1161/atvbaha.116.308895] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 04/25/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Vascular remodeling because of smooth muscle cell (SMC) proliferation is a common process occurring in several vascular diseases, such as atherosclerosis, aortic aneurysm, post-transplant vasculopathy, restenosis after angioplasty, etc. The molecular mechanism underlying SMC proliferation, however, is not completely understood. The objective of this study is to determine the role and mechanism of Janus kinase 3 (JAK3) in vascular remodeling and SMC proliferation. APPROACH AND RESULTS Platelet-derived growth factor-BB, an SMC mitogen, induces JAK3 expression and phosphorylation while stimulating SMC proliferation. Janex-1, a specific inhibitor of JAK3, or knockdown of JAK3 by short hairpin RNA, inhibits the SMC proliferation. Conversely, ectopic expression of JAK3 promotes SMC proliferation. Mechanistically, JAK3 promotes the phosphorylation of signal transducer and activator of transcription 3 and c-Jun N-terminal kinase in SMC, 2 signaling pathways known to be critical for SMC proliferation and vascular remodeling. Blockade of these 2 signaling pathways by their inhibitors impeded the JAK3-mediated SMC proliferation. In vivo, knockdown of JAK3 attenuates injury-induced neointima formation with attenuated neointimal SMC proliferation. Knockdown of JAK3 also induces neointimal SMC apoptosis in rat carotid artery balloon injury model. CONCLUSIONS Our results demonstrate that JAK3 mediates SMC proliferation and survival during injury-induced vascular remodeling, which provides a potential therapeutic target for preventing neointimal hyperplasia in proliferative vascular diseases.
Collapse
Affiliation(s)
- Yung-Chun Wang
- From the Department of Physiology and Pharmacology, University of Georgia, Athens
| | - Xiao-Bing Cui
- From the Department of Physiology and Pharmacology, University of Georgia, Athens
| | - Ya-Hui Chuang
- From the Department of Physiology and Pharmacology, University of Georgia, Athens
| | - Shi-You Chen
- From the Department of Physiology and Pharmacology, University of Georgia, Athens.
| |
Collapse
|
69
|
Lee JJ, Lee JH, Gu MJ, Han JH, Cho WK, Ma JY. Agastache rugosa Kuntze extract, containing the active component rosmarinic acid, prevents atherosclerosis through up-regulation of the cyclin-dependent kinase inhibitors p21WAF1/CIP1 and p27KIP1. J Funct Foods 2017. [DOI: 10.1016/j.jff.2016.12.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
70
|
Pan X, Wang Y, Lübke T, Hinek A, Pshezhetsky AV. Mice, double deficient in lysosomal serine carboxypeptidases Scpep1 and Cathepsin A develop the hyperproliferative vesicular corneal dystrophy and hypertrophic skin thickenings. PLoS One 2017; 12:e0172854. [PMID: 28234994 PMCID: PMC5325571 DOI: 10.1371/journal.pone.0172854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/17/2017] [Indexed: 01/04/2023] Open
Abstract
Vasoactive and mitogenic peptide, endothelin-1 (ET-1) plays an important role in physiology of the ocular tissues by regulating the growth of corneal epithelial cells and maintaining the hemodynamics of intraocular fluids. We have previously established that ET-1 can be degraded in vivo by two lysosomal/secreted serine carboxypeptidases, Cathepsin A (CathA) and Serine Carboxypeptidase 1 (Scpep1) and that gene-targeted CathAS190A/Scpep1-/- mice, deficient in CathA and Scpep1 have a prolonged half-life of circulating ET-1 associated with systemic hypertension. In the current work we report that starting from 6 months of age, ~43% of CathAS190A/Scpep1-/- mice developed corneal clouding that eventually caused vision impairment. Histological evaluation of these mice demonstrated a selective fibrotic thickening and vacuolization of the corneas, resembling human hyperproliferative vesicular corneal stromal dystrophy and coexisting with a peculiar thickening of the skin epidermis. Moreover, we found that cultured corneal epithelial cells, skin fibroblasts and vascular smooth muscle cells derived from CathA/Scpep1-deficient mice, demonstrated a significantly higher proliferative response to treatment with exogenous ET-1, as compared with cells from wild type mice. We also detected increased activation level of ERK1/2 and AKT kinases involved in cell proliferation in the ET-1-treated cultured cells from CathA/Scpep1 deficient mice. Together, results from our experimental model suggest that; in normal tissues the tandem of serine carboxypeptidases, Scpep1 and CathA likely constitutes an important part of the physiological mechanism responsible for the balanced elimination of heightened levels of ET-1 that otherwise would accumulate in tissues and consequently contribute to development of the hyper-proliferative corneal dystrophy and abnormal skin thickening.
Collapse
Affiliation(s)
- Xuefang Pan
- Department of Medical Genetics, CHU Sainte-Justine Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Yanting Wang
- Cardiovascular Research Program, the Hospital for Sick Children and Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Torben Lübke
- Department of Chemistry, Biochemistry I, Bielefeld University, Bielefeld, Germany
| | - Aleksander Hinek
- Cardiovascular Research Program, the Hospital for Sick Children and Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Alexey V. Pshezhetsky
- Department of Medical Genetics, CHU Sainte-Justine Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
71
|
Enhanced Rb/E2F and TSC/mTOR Pathways Induce Synergistic Inhibition in PDGF-Induced Proliferation in Vascular Smooth Muscle Cells. PLoS One 2017; 12:e0170036. [PMID: 28076433 PMCID: PMC5226788 DOI: 10.1371/journal.pone.0170036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/27/2016] [Indexed: 02/04/2023] Open
Abstract
Platelet-derived growth factor (PDGF) plays an essential role in proliferation of vascular smooth muscle cells (VSMCs). The Rb/E2F and TSC/mTOR pathways contribute to the proliferation of VSMCs, but its exact roles in PDGF-induced proliferation are unclear. In this study, we demonstrated the roles of Rb/E2F and TSC/mTOR pathways in PDGF-induced proliferation in VSMCs. We found that PDGF stimulates the activity of E2F and mTOR pathways, and knockdown of either Rb or TSC2 increases PDGF-induced proliferation in VSMCs. More interestingly, we revealed that enhancing both E2F and mTOR activity leads to synergistic inhibition of PDGF-induced proliferation in VSMCs. We further identified that the synergistic inhibition effect is caused by the induced oxidative stress. Summarily, these data suggest the important regulations of Rb/E2F and TSC/mTOR pathways in PDGF-induced proliferation in VSMCs, and also present a promising way to limit deregulated proliferation by PDGF induction in VSMCs.
Collapse
|
72
|
Chen CC, Liang CJ, Leu YL, Chen YL, Wang SH. Viscolin Inhibits In Vitro Smooth Muscle Cell Proliferation and Migration and Neointimal Hyperplasia In Vivo. PLoS One 2016; 11:e0168092. [PMID: 27977759 PMCID: PMC5158191 DOI: 10.1371/journal.pone.0168092] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 11/25/2016] [Indexed: 12/12/2022] Open
Abstract
Viscolin, an extract of Viscum coloratum, has anti-inflammatory and anti-proliferative properties against harmful stimuli. The aim of the study was to examine the anti-proliferative effects of viscolin on platelet derived growth factor-BB (PDGF)-treated human aortic smooth muscle cells (HASMCs) and identify the underlying mechanism responsible for these effects. Viscolin reduced the PDGF-BB-induced HASMC proliferation and migration in vitro; it also arrested HASMCs in the G0/G1 phase by decreasing the protein expression of Cyclin D1, CDK2, Cyclin E, CDK4, and p21Cip1 as detected by Western blot analysis. These effects may be mediated by reduced PDGF-induced phosphorylation of ERK1/2, JNK, and P38, but not AKT as well as inhibition of PDGF-mediated nuclear factor (NF)-κB p65 and activator protein 1 (AP-1)/c-fos activation. Furthermore, viscolin pre-treatment significantly reduced neointimal hyperplasia of an endothelial-denuded femoral artery in vivo. Taken together, viscolin attenuated PDGF–BB-induced HASMC proliferation in vitro and reduced neointimal hyperplasia in vivo. Thus, viscolin may represent a therapeutic candidate for the prevention and treatment of vascular proliferative diseases.
Collapse
Affiliation(s)
- Chin-Chuan Chen
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chan-Jung Liang
- Center for Lipid and Glycomedicine Research (CLGR), Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Lipid Biosciences (CLB), Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
- Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Huei Wang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
73
|
Kim GY, Park JH, Kim H, Lim HJ, Park HY. Coronin 1B serine 2 phosphorylation by p38α is critical for vascular endothelial growth factor-induced migration of human umbilical vein endothelial cells. Cell Signal 2016; 28:1817-1825. [DOI: 10.1016/j.cellsig.2016.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/06/2016] [Accepted: 08/15/2016] [Indexed: 11/30/2022]
|
74
|
Ge X, Chen SY, Liu M, Liang TM, Liu C. Evodiamine inhibits PDGF‑BB‑induced proliferation of rat vascular smooth muscle cells through the suppression of cell cycle progression and oxidative stress. Mol Med Rep 2016; 14:4551-4558. [PMID: 27748810 PMCID: PMC5101993 DOI: 10.3892/mmr.2016.5798] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 09/19/2016] [Indexed: 12/20/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) proliferation is a key event in the development of in-stent restenosis. Evodiamine is an indole alkaloid extracted from the Chinese medicine, evodia, and has been shown to inhibit tumor cell proliferation and protect the cardiovascular system. However, whether evodiamine affects VSMC proliferation remains to be elucidated. Therefore, the present study examined the effects and the mechanisms of action of evodiamine on the proliferation of rat VSMCs. The cells were treated with evodiamine alone or in combination with platelet-derived growth factor-BB (PDGF-BB) stimulation. It was found that evodiamine inhibited PDGF-BB-induced VSMC proliferation in a dose-dependent manner, without inducing cell death. Evodiamine also retarded cell cycle progression, evidenced by the suppression of the expression of cell cycle-promoting cyclin proteins and cyclin-dependent kinases. In addition, evodiamine attenuated the PDGF-BB-induced phosphorylation of mitogen-activated protein kinases p38 and extracellular signal-regulated kinases 1/2, however, it had no effect on the phosphorylation of Akt. Evodiamine also inhibited the increase of reactive oxygen species generation and upregulated the mRNA expression levels of genes encoding antioxidant enzymes. These findings provide important insights into the mechanisms underlying the vasoprotective actions of evodiamine and suggest that it may be a useful therapeutic agent for the treatment of vascular occlusive disease.
Collapse
Affiliation(s)
- Xie Ge
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology and College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Si-Yu Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology and College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Mei Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology and College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Ting-Ming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology and College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Chang Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology and College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
75
|
Pandey P, Ali Z, Mohammad G, Pasha MAQ. Elevated blood plasma levels of epinephrine, norepinephrine, tyrosine hydroxylase, TGFβ1, and TNFα associated with high-altitude pulmonary edema in an Indian population. Ther Clin Risk Manag 2016; 12:1207-21. [PMID: 27540296 PMCID: PMC4982497 DOI: 10.2147/tcrm.s111030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Biomarkers are essential to unravel the locked pathophysiology of any disease. This study investigated the role of biomarkers and their interactions with each other and with the clinical parameters to study the physiology of high-altitude pulmonary edema (HAPE) in HAPE-patients (HAPE-p) against adapted highlanders (HLs) and healthy sojourners, HAPE-controls (HAPE-c). For this, seven circulatory biomarkers, namely, epinephrine, norepinephrine, tyrosine hydroxylase, transforming growth factor beta 1, tumor necrosis factor alpha (TNFα), platelet-derived growth factor beta beta, and C-reactive protein (CRP), were measured in blood plasma of the three study groups. All the subjects were recruited at ~3,500 m, and clinical features such as arterial oxygen saturation (SaO2), body mass index, and mean arterial pressure were measured. Increased levels of epinephrine, norepinephrine, tyrosine hydroxylase, transforming growth factor-beta 1, and TNFα were observed in HAPE-p against the healthy groups, HAPE-c, and HLs (P<0.0001). CRP levels were decreased in HAPE-p against HAPE-c and HLs (P<0.0001). There was no significant difference or very marginal difference in the levels of these biomarkers in HAPE-c and HLs (P>0.01). Correlation analysis revealed a negative correlation between epinephrine and norepinephrine (P=4.6E−06) in HAPE-p and positive correlation in HAPE-c (P=0.004) and HLs (P=9.78E−07). A positive correlation was observed between TNFα and CRP (P=0.004) in HAPE-p and a negative correlation in HAPE-c (P=4.6E−06). SaO2 correlated negatively with platelet-derived growth factor beta beta (HAPE-p; P=0.05), norepinephrine (P=0.01), and TNFα (P=0.005) and positively with CRP (HAPE-c; P=0.02) and norepinephrine (HLs; P=0.04). Body mass index correlated negatively with epinephrine (HAPE-p; P=0.001) and positively with norepinephrine and tyrosine hydroxylase in HAPE-c (P<0.05). Mean arterial pressure correlated positively with TNFα in HAPE-p and norepinephrine in HLs (P<0.05). Receiver operating characteristic curve analysis yielded a positive predictive value for these biomarkers with HAPE (area under the curve >0.70, P<0.05). The results clearly suggest that increased plasma levels of these circulatory biomarkers associated with HAPE.
Collapse
Affiliation(s)
- Priyanka Pandey
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi; Department of Biotechnology, Savitribai Phule Pune University, Pune
| | - Zahara Ali
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi; Department of Biotechnology, Savitribai Phule Pune University, Pune
| | - Ghulam Mohammad
- Department of Medicine, SNM Hospital, Ladakh, Jammu and Kashmir, India
| | - M A Qadar Pasha
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi; Department of Biotechnology, Savitribai Phule Pune University, Pune
| |
Collapse
|
76
|
Lee JJ, Lee JH, Cho WK, Han JH, Ma JY. Herbal composition of Cinnamomum cassia, Pinus densiflora, Curcuma longa and Glycyrrhiza glabra prevents atherosclerosis by upregulating p27 (Kip1) expression. Altern Ther Health Med 2016; 16:253. [PMID: 27465365 PMCID: PMC4964310 DOI: 10.1186/s12906-016-1224-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 07/15/2016] [Indexed: 12/30/2022]
Abstract
Background Kiom-18 is a novel composition of Cinnamomum cassia, Pinus densiflora, Curcuma longa and Glycyrrhiza glabra. Curcuma longa and Glycyrrhiza glabra, which are traditional medicines in Asia, have been reported to demonstrate preventive effects against atherosclerosis; however, they have not yet been developed into functional atherosclerosis treatments. We therefore studied the anti-atherosclerotic effects and possible molecular mechanisms of Kiom-18 using vascular smooth muscle cells (VSMCs). Methods To assess the anti-proliferative effect of Kiom-18 in vitro, we performed thymidine incorporation, cell cycle progression, immunoblotting and immunofluorescence assays in VSMCs stimulated by platelet derived-growth factor (PDGF)-BB. In addition, we used LDLr knockout mice to identify the effects of Kiom-18 as a preliminary result in an atherosclerosis animal model. Results Kiom-18 inhibited platelet-derived growth factor (PDGF)-BB-stimulated-VSMC proliferation and DNA synthesis. Additionally, Kiom-18 arrested the cell cycle transition of G0/G1 stimulated by PDGF-BB and its cell cycle-related proteins. Correspondingly, the level of p27kip1 expression was upregulated in the presence of the Kiom-18 extract. Moreover, in an atherosclerosis animal model of LDLr knockout mice, Kiom-18 extract showed a preventive effect for the formation of atherosclerotic plaque and suppressed body weight, fat weight, food treatment efficiency, neutrophil count, and triglyceride level. Conclusions These results indicate that Kiom-18 exerts anti-atherosclerotic effects by inhibiting VSMC proliferation via G0/G1 arrest, which upregulates p27Kip1 expression.
Collapse
|
77
|
Irisin reverses platelet derived growth factor-BB-induced vascular smooth muscle cells phenotype modulation through STAT3 signaling pathway. Biochem Biophys Res Commun 2016; 479:139-145. [PMID: 27416763 DOI: 10.1016/j.bbrc.2016.07.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/09/2016] [Indexed: 01/16/2023]
Abstract
Vascular smooth muscle cells (VSMCs) phenotype modulation toward a synthetic phenotype is the main cause of cardiovascular disease. As a newly discovered myokine, Irisin is thought to be a promising candidate for the treatment of metabolic disturbances, as well as cardiovascular disease. However, no evidence has been shown for the direct effect of Irisin on VSMCs phenotype modulation and its underling mechanisms. The aim of this study was to explore the effect of Irisin on VSMCs phenotype modulation and the mechanisms involved. In the present study, it was found that Irisin restored the PDGF-BB-induced VSMCs phenotype modulation which exhibited down-regulation of smooth muscle cells (SMC) expression and up-regulation of matrix synthesis related marker expression, as well as proliferative phenotype. Moreover, our research demonstrated that Irisin further activated STAT3 signaling pathways. Finally, by applying an STAT3 inhibitor, WP1066, we revealed the roles of STAT3 in the PDGF-BB-induced VSMCs phenotype modulation when they were treated with Irisin. Taken together, these results demonstrated that Irisin may play a crucial role in regulating VSMCs phenotype modulation via the STAT3 signaling pathway.
Collapse
|
78
|
Shh mediates PDGF-induced contractile-to-synthetic phenotypic modulation in vascular smooth muscle cells through regulation of KLF4. Exp Cell Res 2016; 345:82-92. [DOI: 10.1016/j.yexcr.2016.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/11/2016] [Accepted: 05/15/2016] [Indexed: 12/31/2022]
|
79
|
Singh R. Model Predicts That MKP1 and TAB1 Regulate p38α Nuclear Pulse and Its Basal Activity through Positive and Negative Feedback Loops in Response to IL-1. PLoS One 2016; 11:e0157572. [PMID: 27314954 PMCID: PMC4912083 DOI: 10.1371/journal.pone.0157572] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/01/2016] [Indexed: 01/20/2023] Open
Abstract
Interleukin-1 mediates inflammation and stress response through nuclear activity of p38α. Although IL-1 receptor is not degraded, p38α activation is transient. IL-1 also causes cell migration and EMT by modulating cell-cell junctions. Although molecules involved in p38 activation are known, mechanism of the transient nuclear response and its basal activity remains unknown. By mathematical modeling of IL1/p38 signaling network, we show that IL-1 induces robust p38α activation both in the nucleus and in the cytoplasm/membrane. While nuclear response consists of an acute phase, membrane response resembles a step change. Following stimulation, p38α activity returns to a basal level in absence of receptor degradation. While nuclear pulse is controlled by MKP1 through a negative feedback to pp38, its basal activity is controlled by both TAB1 and MKP1 through a positive feedback loop. Our model provides insight into the mechanism of p38α activation, reason for its transient nuclear response, and explanation of the basal activity of MKK3/6 and p38α, which has been experimentally observed by other groups.
Collapse
Affiliation(s)
- Raghvendra Singh
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, India
- * E-mail:
| |
Collapse
|
80
|
Magnobovatol inhibits smooth muscle cell migration by suppressing PDGF-Rβ phosphorylation and inhibiting matrix metalloproteinase-2 expression. Int J Mol Med 2016; 37:1239-46. [PMID: 27049716 PMCID: PMC4829143 DOI: 10.3892/ijmm.2016.2548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/29/2016] [Indexed: 11/23/2022] Open
Abstract
The migration of vascular smooth muscle cells (VSMCs) may play a crucial role in the pathogenesis of vascular diseases, such as atherosclerosis and post-angioplasty restenosis. Platelet-derived growth factor (PDGF)-BB is a potent mitogen for VSMCs and plays an important role in the intimal accumulation of VSMCs. Magnobovatol, a new neolignan from the fruits of Magnolia obovata, has been shown to have anticancer properties. However, the effects of magnobovatol on VSMCs are unknown. In the present study, we examined the effects of magnobovatol on the PDGF-BB-induced migration of mouse and human VSMCs, as well as the underlying mechanisms. Magnobovatol significantly inhibited the PDGF-BB-induced migration of mouse and human VSMCs without inducing cell death (as shown by MTT assay and wound healing assay). Additionally, we demonstrated that magnobovatol significantly blocked the PDGF-BB-induced phosphorylation of the PDGF receptor (PDGF-R), Akt and extracellular signal-regulated kinase (ERK)1/2 by inhibiting the activation of the PDGF-BB signaling pathway. Moreover, in both mouse and human VSMCs, magnobovatol inhibited PDGF-induced matrix metalloproteinase (MMP)-2 expression at the mRNA and protein level, as well as the proteolytic activity of MMP-2 (as shown by western blot analysis, RT-PCR, gelatin zymography and ELISA). In addition, the sprout outgrowth formation of aortic rings induced by PDGF-BB was inhibited by magnobovatol (as shown by aortic ring assay). Taken together, our findings indicate that magnobovatol inhibits VSMC migration by decreasing MMP-2 expression through PDGF-R and the ERK1/2 and Akt pathways. Our data may improve the understanding of the anti-atherogenic effects of magnobovatol in VSMCs.
Collapse
|
81
|
Ganguly R, Wen AM, Myer AB, Czech T, Sahu S, Steinmetz NF, Raman P. Anti-atherogenic effect of trivalent chromium-loaded CPMV nanoparticles in human aortic smooth muscle cells under hyperglycemic conditions in vitro. NANOSCALE 2016; 8:6542-6554. [PMID: 26935414 PMCID: PMC5136293 DOI: 10.1039/c6nr00398b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Atherosclerosis, a major macrovascular complication associated with diabetes, poses a tremendous burden on national health care expenditure. Despite extensive efforts, cost-effective remedies are unknown. Therapies for atherosclerosis are challenged by a lack of targeted drug delivery approaches. Toward this goal, we turn to a biology-derived drug delivery system utilizing nanoparticles formed by the plant virus, Cowpea mosaic virus (CPMV). The aim herein is to investigate the anti-atherogenic potential of the beneficial mineral nutrient, trivalent chromium, loaded CPMV nanoparticles in human aortic smooth muscle cells (HASMC) under hyperglycemic conditions. A non-covalent loading protocol is established yielding CrCl3-loaded CPMV (CPMV-Cr) carrying 2000 drug molecules per particle. Using immunofluorescence microscopy, we show that CPMV-Cr is readily taken up by HASMC in vitro. In glucose (25 mM)-stimulated cells, 100 nM CPMV-Cr inhibits HASMC proliferation concomitant to attenuated proliferating cell nuclear antigen (PCNA, proliferation marker) expression. This is accompanied by attenuation in high glucose-induced phospho-p38 and pAkt expression. Moreover, CPMV-Cr inhibits the expression of pro-inflammatory cytokines, transforming growth factor-β (TGF-β) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), in glucose-stimulated HASMCs. Finally glucose-stimulated lipid uptake is remarkably abrogated by CPMV-Cr, revealed by Oil Red O staining. Together, these data provide key cellular evidence for an atheroprotective effect of CPMV-Cr in vascular smooth muscle cells (VSMC) under hyperglycemic conditions that may promote novel therapeutic ventures for diabetic atherosclerosis.
Collapse
Affiliation(s)
- Rituparna Ganguly
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272-0095, USA. and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Amy M Wen
- Department of Biomedical Engineering, 10990 Euclid Avenue and Case Western Reserve University, Cleveland, OH, USA
| | - Ashley B Myer
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272-0095, USA.
| | - Tori Czech
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272-0095, USA.
| | - Soumyadip Sahu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272-0095, USA. and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, 10990 Euclid Avenue and Case Western Reserve University, Cleveland, OH, USA and Department of Radiology, 10990 Euclid Avenue and Case Western Reserve University, Cleveland, OH, USA and Department of Materials Science and Engineering, 10990 Euclid Avenue and Case Western Reserve University, Cleveland, OH, USA and Department of Macromolecular Science and Engineering, 10990 Euclid Avenue and Case Western Reserve University, Cleveland, OH, USA and Case Comprehensive Cancer Center, 10990 Euclid Avenue and Case Western Reserve University, Cleveland, OH, USA
| | - Priya Raman
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272-0095, USA. and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
82
|
Shimizu H, Hagio M, Iwaya H, Tsuneki I, Lee JY, Fukiya S, Yokota A, Miyazaki H, Hara H, Ishizuka S. Deoxycholic acid is involved in the proliferation and migration of vascular smooth muscle cells. J Nutr Sci Vitaminol (Tokyo) 2016; 60:450-4. [PMID: 25866311 DOI: 10.3177/jnsv.60.450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Obesity is increasingly becoming associated with increased risk of atherosclerosis. Serum levels of the bile acid deoxycholic acid (DCA) are elevated in mice with obesity induced by a high-fat (HF) diet. Therefore, we investigated the influence of DCA on the functions of vascular smooth muscle cells (VSMCs) because the initiation and progression of atherosclerosis are associated with VSMC proliferation and migration. DCA induced c-jun N-terminal kinase (JNK) activation whereas a JNK inhibitor prevented DCA-induced VSMC proliferation and migration. Based on these findings, we examined whether DCA promotes the expression of platelet-derived growth factor β-receptor (PDGFRβ) that has a c-Jun binding site in its promoter region. The mRNA and protein expression levels of PDGFRβ were upregulated in VSMCs after a 24- and 48-h incubation with DCA, respectively. The effects of PDGF such as proliferation and migration of VSMCs were promoted after a 48-h incubation with DCA despite the absence of DCA during PDGF stimulation. These findings suggest that elevated serum concentrations of DCA are involved in the pathogenesis of atherosclerosis in HF-induced obesity.
Collapse
Affiliation(s)
- Hidehisa Shimizu
- Division of Applied Bioscience, Research Faculty of Agriculture, Hokkaido University
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
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: 0.7] [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
|
84
|
Glucocorticoid-induced fetal origins of adult hypertension: Association with epigenetic events. Vascul Pharmacol 2016; 82:41-50. [PMID: 26903240 DOI: 10.1016/j.vph.2016.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 02/05/2023]
Abstract
Hypertension is a predominant risk factor for cardiovascular diseases and a major health care burden. Accumulating epidemiological and experimental evidence suggest that adult-onset hypertension may have its origins during early development. Upon exposure to glucocorticoids, the fetus develops hypertension, and the offspring may be programmed to continue the hypertensive trajectory into adulthood. Elevated oxidative stress and deranged nitric oxide system are not only hallmarks of adult hypertension but are also observed earlier in life. Endothelial dysfunction and remodeling of the vasculature, which are robustly associated with increased incidence of hypertension, are likely to have been pre-programmed during fetal life. Apparently, genomic, non-genomic, and epigenomic factors play a significant role in the development of hypertension, including glucocorticoid-driven effects on blood pressure. In this review, we discuss the involvement of the aforementioned participants in the pathophysiology of hypertension and suggest therapeutic opportunities for targeting epigenome modifiers, potentially for personalized medicine.
Collapse
|
85
|
Sun Q, Liu L, Mandal J, Molino A, Stolz D, Tamm M, Lu S, Roth M. PDGF-BB induces PRMT1 expression through ERK1/2 dependent STAT1 activation and regulates remodeling in primary human lung fibroblasts. Cell Signal 2016; 28:307-15. [PMID: 26795953 DOI: 10.1016/j.cellsig.2016.01.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/11/2016] [Accepted: 01/15/2016] [Indexed: 12/16/2022]
Abstract
Tissue remodeling of sub-epithelial mesenchymal cells is a major pathology occurring in chronic obstructive pulmonary disease (COPD) and asthma. Fibroblasts, as a major source of interstitial connective tissue extracellular matrix, contribute to the fibrotic and inflammatory changes in these airways diseases. Previously, we described that protein arginine methyltransferase-1 (PRMT1) participates in airway remodeling in a rat model of pulmonary inflammation. In this study we investigated the mechanism by which PDGF-BB regulates PRMT1 in primary lung fibroblasts, isolated from human lung biopsies. Fibroblasts were stimulated with PDGF-BB for up-to 48h and the regulatory and activation of signaling pathways controlling PRMT1 expression were determined. PRMT1 was localized by immuno-histochemistry in human lung tissue sections and by immunofluorescence in isolated fibroblasts. PRMT1 activity was suppressed by the pan-PRMT inhibitor AMI1. ERK1/2 mitogen activated protein kinase (MAPK) was blocked by PD98059, p38 MAPK by SB203580, and STAT1 by small interference (si) RNA treatment. The results showed that PDGF-BB significantly increased PRMT1 expression after 1h lasting over 48h, through ERK1/2 MAPK and STAT1 signaling. The inhibition of ERK1/2 MAPK or of PRMT1 activity decreased PDGF-BB induced fibroblast proliferation, COX2 production, collagen-1A1 secretion, and fibronectin production. These findings suggest that PRMT1 is a central regulator of tissue remodeling and that the signaling sequence controlling its expression in primary human lung fibroblast is PDGF-ERK-STAT1. Therefore, PRMT1 presents a novel therapeutic and diagnostic target for the control of airway wall remodeling in chronic lung diseases.
Collapse
Affiliation(s)
- Qingzhu Sun
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Pneumology and Pulmonary Cell Research, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
| | - Li Liu
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China
| | - Jyotshna Mandal
- Pneumology and Pulmonary Cell Research, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
| | - Antonio Molino
- Dept of Respiratory Diseases, University of Naples, Federico II, Naples, Italy
| | - Daiana Stolz
- Pneumology and Pulmonary Cell Research, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
| | - Michael Tamm
- Pneumology and Pulmonary Cell Research, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China
| | - Michael Roth
- Pneumology and Pulmonary Cell Research, Department of Biomedicine, University and University Hospital Basel, Basel 4031, Switzerland.
| |
Collapse
|
86
|
Daniel JM, Prock A, Dutzmann J, Sonnenschein K, Thum T, Bauersachs J, Sedding DG. Regulator of G-Protein Signaling 5 Prevents Smooth Muscle Cell Proliferation and Attenuates Neointima Formation. Arterioscler Thromb Vasc Biol 2015; 36:317-27. [PMID: 26663397 DOI: 10.1161/atvbaha.115.305974] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/24/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Regulator of G-protein signaling 5 (RGS5) is abundantly expressed in vascular smooth muscle cells (SMCs) and inhibits G-protein signaling by enhancing the guanosine triphosphate-hydrolyzing activity of Gα-subunits. In the present study, we investigated the effects of RGS5 on vascular SMC function in vitro and neointima formation after wire-induced injury in mice and determined the underlying mechanisms. APPROACH AND RESULTS We found a robust expression of RGS5 in native arteries of C57BL/6 mice and a highly significant downregulation within neointimal lesions 10 and 21 days after vascular injury as assessed by quantitative polymerase chain reaction, immunoblotting, and immunohistochemistry. In vitro, RGS5 was found significantly downregulated after mitogenic stimulation of human coronary artery SMCs. To restore RGS5 levels, SMCs were transduced with adenoviral vectors encoding wild-type RGS5 or a nondegradable mutant. RGS5-WT and, even more prominently, the C2A-RGS5 mutant prevented SMC proliferation and migration. In contrast, the siRNA-mediated knockdown of RGS5 significantly augmented SMC proliferation. Following overexpression of RGS5, fluorescence-activated cell sorting analysis of propidium iodide-stained cells indicated cell cycle arrest in G0/G1 phase. Mechanistically, inhibition of the phosphorylation of the extracellular signal-regulated kinase 1/2 and mitogen-activated protein kinase downstream signaling was shown to be responsible for the anti-proliferative effect of RGS5. Following wire-induced injury of the femoral artery in C57BL/6 mice, adenoviral-mediated overexpression of RGS5-WT or C2A-RGS5 significantly reduced SMC proliferation and neointima formation in vivo. CONCLUSIONS Downregulation of RGS5 is an important prerequisite for SMC proliferation in vitro and in vivo. Therefore, reconstitution of RGS5 levels represents a promising therapeutic option to prevent vascular remodeling processes.
Collapse
Affiliation(s)
- Jan-Marcus Daniel
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - André Prock
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Jochen Dutzmann
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Kristina Sonnenschein
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Thomas Thum
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Johann Bauersachs
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.)
| | - Daniel G Sedding
- From the Department of Cardiology and Angiology (J.-M.D., J.D., K.S., J.B., D.G.S.), REBIRTH Excellence Cluster (J.-M.D., T.T., J.B., D.G.S.), and Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (K.S., T.T.), Hannover Medical School, Hannover, Germany; Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany (A.P.); and National Heart and Lung Institute, Imperial College London, London, UK (T.T.).
| |
Collapse
|
87
|
Schlosser A, Pilecki B, Hemstra LE, Kejling K, Kristmannsdottir GB, Wulf-Johansson H, Moeller JB, Füchtbauer EM, Nielsen O, Kirketerp-Møller K, Dubey LK, Hansen PBL, Stubbe J, Wrede C, Hegermann J, Ochs M, Rathkolb B, Schrewe A, Bekeredjian R, Wolf E, Gailus-Durner V, Fuchs H, Hrabě de Angelis M, Lindholt JS, Holmskov U, Sorensen GL. MFAP4 Promotes Vascular Smooth Muscle Migration, Proliferation and Accelerates Neointima Formation. Arterioscler Thromb Vasc Biol 2015; 36:122-33. [PMID: 26564819 DOI: 10.1161/atvbaha.115.306672] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/29/2015] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Arterial injury stimulates remodeling responses that, when excessive, lead to stenosis. These responses are influenced by integrin signaling in vascular smooth muscle cells (VSMCs). Microfibrillar-associated protein 4 (MFAP4) is an integrin ligand localized to extracellular matrix fibers in the vascular wall. The role of MFAP4 in vascular biology is unknown. We aimed to test the hypothesis that MFAP4 would enhance integrin-dependent VSMC activation. APPROACH AND RESULTS We produced Mfap4-deficient (Mfap4(-/-)) mice and performed carotid artery ligation to explore the role of MFAP4 in vascular biology in vivo. Furthermore, we investigated the effects of MFAP4 in neointimal formation ex vivo and in primary VSMC and monocyte cultures in vitro. When challenged with carotid artery ligation, Mfap4(-/-) mice exhibited delayed neointimal formation, accompanied by early reduction in the number of proliferating medial and neointimal cells, as well as infiltrating leukocytes. Delayed neointimal formation was associated with decreased cross-sectional area of ligated Mfap4(-/-) carotid arteries resulting in lumen narrowing 28 days after ligation. MFAP4 blockade prohibited the formation of neointimal hyperplasia ex vivo. Moreover, we demonstrated that MFAP4 is a ligand for integrin αVβ3 and mediates VSMC phosphorylation of focal adhesion kinase, migration, and proliferation in vitro. MFAP4-dependent VSMC activation was reversible by treatment with MFAP4-blocking antibodies and inhibitors of focal adhesion kinase and downstream kinases. In addition, we showed that MFAP4 promotes monocyte chemotaxis in integrin αVβ3-dependent manner. CONCLUSIONS MFAP4 regulates integrin αVβ3-induced VSMC proliferation and migration, as well as monocyte chemotaxis, and accelerates neointimal hyperplasia after vascular injury.
Collapse
Affiliation(s)
- Anders Schlosser
- From the Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark (A.S., B.P., L.E.H., K.K., G.B.K., H.W.-J., J.B.M., K.K.-M., L.K.D., P.B.L.H., J.S., U.H, G.L.S.); Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (E.-M.F.); Department of Pathology, Odense University Hospital, Odense, Denmark (O.N.); Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany (C.W., J.H., M.O.); Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany (C.W., J.H., M.O.); REBIRTH Cluster of Excellence, Hannover, Germany (C.W., J.H., M.O.); German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany (B.R., A.S., V.G.-D., H.F., M.H.d.A.); Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Munich, Germany (B.R, E.W.); Division of Cardiology, Department of Medicine III, University of Heidelberg, Heidelberg, Germany (A.S., R.B.); Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Munich, Germany (M.H.d.A.); Cardiovascular Research Unit, Viborg Hospital, Viborg, Denmark (J.S.L.); and Department of Cardiothoracic and Vascular Surgery, Center of Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark (J.S.L.)
| | - Bartosz Pilecki
- From the Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark (A.S., B.P., L.E.H., K.K., G.B.K., H.W.-J., J.B.M., K.K.-M., L.K.D., P.B.L.H., J.S., U.H, G.L.S.); Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark (E.-M.F.); Department of Pathology, Odense University Hospital, Odense, Denmark (O.N.); Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany (C.W., J.H., M.O.); Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany (C.W., J.H., M.O.); REBIRTH Cluster of Excellence, Hannover, Germany (C.W., J.H., M.O.); German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany (B.R., A.S., V.G.-D., H.F., M.H.d.A.); Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Munich, Germany (B.R, E.W.); Division of Cardiology, Department of Medicine III, University of Heidelberg, Heidelberg, Germany (A.S., R.B.); Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Munich, Germany (M.H.d.A.); Cardiovascular Research Unit, Viborg Hospital, Viborg, Denmark (J.S.L.); and Department of Cardiothoracic and Vascular Surgery, Center of Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark (J.S.L.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
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: 2.0] [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
|
89
|
Song JH, Jeong GH, Park SL, Won SY, Paek NS, Lee BH, Moon SK. Inhibitory effects of fermented extract of Ophiopogon japonicas on thrombin-induced vascular smooth muscle cells. Mol Med Rep 2015; 13:426-32. [PMID: 26530246 DOI: 10.3892/mmr.2015.4499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 09/03/2015] [Indexed: 11/06/2022] Open
Abstract
Ophiopogon japonicus is known to have various pharmacological effects. The present study investigated the effects of an extract of fermented Ophiopogon japonicas (FEOJ) on thrombin‑treated vascular smooth muscle cells (VSMCs). FEOJ treatment inhibited the proliferation of VSMCs treated with thrombin as indicated by an MTT assay. These inhibitory effects were associated with decreased phosphorylation of AKT, reduced expression of cyclin D1 and increased expression of p27KIP1 in thrombin‑induced VSMCs. In addition, FEOJ treatment suppressed the thrombin‑stimulated migration of VSMCs as demonstrated by a wound‑healing migration assay. Furthermore, zymographic analyses demonstrated that treatment of FEOJ with VSMCs suppressed the thrombin‑induced expression of matrix metalloproteinase (MMP)‑2, which was attributed to the reduction of nuclear factor (NF)‑κB binding activity. Collectively, these results demonstrated that FEOJ induced p27KIP1 expression, reduced cyclin D1 expression and AKT phosphorylation, and inhibited MMP‑2 expression mediated by downregulation of NF‑κB binding activity in thrombin‑treated VSMCs, which led to growth inhibition and repression of migration. These results supported the use of FEOJ for the prevention of vascular diseases and provided novel insight into the underlying mechanism of action.
Collapse
Affiliation(s)
- Jun-Hui Song
- Department of Food and Nutrition, Chung‑Ang University, Anseong‑si, Gyeonggi‑do 456‑756, Republic of Korea
| | - Gi Hee Jeong
- Department of Food and Nutrition, Chung‑Ang University, Anseong‑si, Gyeonggi‑do 456‑756, Republic of Korea
| | - Sung Lyea Park
- Department of Food and Nutrition, Chung‑Ang University, Anseong‑si, Gyeonggi‑do 456‑756, Republic of Korea
| | - Se Yeon Won
- Department of Food and Nutrition, Chung‑Ang University, Anseong‑si, Gyeonggi‑do 456‑756, Republic of Korea
| | - Nam Soo Paek
- Mediogen, Jecheon‑si, Chungcheongbuk‑do 390‑250, Republic of Korea
| | - Bog-Hieu Lee
- Department of Food and Nutrition, Chung‑Ang University, Anseong‑si, Gyeonggi‑do 456‑756, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung‑Ang University, Anseong‑si, Gyeonggi‑do 456‑756, Republic of Korea
| |
Collapse
|
90
|
Cohen DPA, Martignetti L, Robine S, Barillot E, Zinovyev A, Calzone L. Mathematical Modelling of Molecular Pathways Enabling Tumour Cell Invasion and Migration. PLoS Comput Biol 2015; 11:e1004571. [PMID: 26528548 PMCID: PMC4631357 DOI: 10.1371/journal.pcbi.1004571] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 09/29/2015] [Indexed: 02/07/2023] Open
Abstract
Understanding the etiology of metastasis is very important in clinical perspective, since it is estimated that metastasis accounts for 90% of cancer patient mortality. Metastasis results from a sequence of multiple steps including invasion and migration. The early stages of metastasis are tightly controlled in normal cells and can be drastically affected by malignant mutations; therefore, they might constitute the principal determinants of the overall metastatic rate even if the later stages take long to occur. To elucidate the role of individual mutations or their combinations affecting the metastatic development, a logical model has been constructed that recapitulates published experimental results of known gene perturbations on local invasion and migration processes, and predict the effect of not yet experimentally assessed mutations. The model has been validated using experimental data on transcriptome dynamics following TGF-β-dependent induction of Epithelial to Mesenchymal Transition in lung cancer cell lines. A method to associate gene expression profiles with different stable state solutions of the logical model has been developed for that purpose. In addition, we have systematically predicted alleviating (masking) and synergistic pairwise genetic interactions between the genes composing the model with respect to the probability of acquiring the metastatic phenotype. We focused on several unexpected synergistic genetic interactions leading to theoretically very high metastasis probability. Among them, the synergistic combination of Notch overexpression and p53 deletion shows one of the strongest effects, which is in agreement with a recent published experiment in a mouse model of gut cancer. The mathematical model can recapitulate experimental mutations in both cell line and mouse models. Furthermore, the model predicts new gene perturbations that affect the early steps of metastasis underlying potential intervention points for innovative therapeutic strategies in oncology.
Collapse
Affiliation(s)
- David P. A. Cohen
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Loredana Martignetti
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Sylvie Robine
- Institut Curie, Paris, France
- CNRS UMR144, Paris, France
| | - Emmanuel Barillot
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Andrei Zinovyev
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Laurence Calzone
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
- * E-mail:
| |
Collapse
|
91
|
Abraham NG, Junge JM, Drummond GS. Translational Significance of Heme Oxygenase in Obesity and Metabolic Syndrome. Trends Pharmacol Sci 2015; 37:17-36. [PMID: 26515032 DOI: 10.1016/j.tips.2015.09.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/10/2015] [Accepted: 09/17/2015] [Indexed: 01/04/2023]
Abstract
The global epidemic of obesity continues unabated with sequelae of diabetes and metabolic syndrome. This review reflects the dramatic increase in research on the role of increased expression of heme oxygenase (HO)-1/HO-2, biliverdin reductase, and HO activity on vascular disease. The HO system engages with other systems to mitigate the deleterious effects of oxidative stress in obesity and cardiovascular disease (CVD). Recent reports indicate that HO-1/HO-2 protein expression and HO activity have several important roles in hemostasis and reactive oxygen species (ROS)-dependent perturbations associated with metabolic syndrome. HO-1 protects tissue during inflammatory stress in obesity through the degradation of pro-oxidant heme and the production of carbon monoxide (CO) and bilirubin, both of which have anti-inflammatory and anti-apoptotic properties. By contrast, repression of HO-1 is associated with increases of cellular heme and inflammatory conditions including hypertension, stroke, and atherosclerosis. HO-1 is a major focus in the development of potential therapeutic strategies to reverse the clinical complications of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA; Marshall University, Joan C. Edwards School of Medicine, Huntington, WV 25701, USA.
| | - Joshua M Junge
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA
| | - George S Drummond
- Departments of Medicine and Pharmacology, New York Medical College, School of Medicine, Valhalla, NY 10595, USA
| |
Collapse
|
92
|
Lee CJ, Lee MH, Yoo SM, Choi KI, Song JH, Jang JH, Oh SR, Ryu HW, Lee HS, Surh YJ, Cho YY. Magnolin inhibits cell migration and invasion by targeting the ERKs/RSK2 signaling pathway. BMC Cancer 2015; 15:576. [PMID: 26253302 PMCID: PMC4529708 DOI: 10.1186/s12885-015-1580-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 07/27/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Magnolin is a natural compound abundantly found in Magnolia flos, which has been traditionally used in oriental medicine to treat headaches, nasal congestion and anti-inflammatory reactions. Our recent results have demonstrated that magnolin targets the active pockets of ERK1 and ERK2, which are important signaling molecules in cancer cell metastasis. The aim of this study is to evaluate the effects of magnolin on cell migration and to further explore the molecular mechanisms involved. METHODS Magnolin-mediated signaling inhibition was confirmed by Western blotting using RSK2(+/+) and RSK2(-/-) MEFs, A549 and NCI-H1975 lung cancer cells, and by NF-κB and Cox-2 promoter luciferase reporter assays. Inhibition of cell migration by magnolin was examined by wound healing and/or Boyden Chamber assays using JB6 Cl41 and A549 human lung cancer cells. The molecular mechanisms involved in cell migration and epithelial-to-mesenchymal transition were determined by zymography, Western blotting, real-time PCR and immunocytofluorescence. RESULTS Magnolin inhibited NF-κB transactivation activity by suppressing the ERKs/RSK2 signaling pathway. Moreover, magnolin abrogated the increase in EGF-induced COX-2 protein levels and wound healing. In human lung cancer cells such as A549 and NCI-H1975, which harbor constitutive active Ras and EGFR mutants, respectively, magnolin suppressed wound healing and cell invasion as seen by a Boyden chamber assay. In addition, it was observed that magnolin inhibited MMP-2 and -9 gene expression and activity. The knockdown or knockout of RSK2 in A549 lung cancer cells or MEFs revealed that magnolin targeting ERKs/RSK2 signaling suppressed epithelial-to-mesenchymal transition by modulating EMT marker proteins such as N-cadherin, E-cadherin, Snail, Vimentin and MMPs. CONCLUSIONS These results demonstrate that magnolin inhibits cell migration and invasion by targeting the ERKs/RSK2 signaling pathway.
Collapse
Affiliation(s)
- Cheol-Jung Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Mee-Hyun Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Sun-Mi Yoo
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Kyung-Il Choi
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Ji-Hong Song
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Jeong-Hoon Jang
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea. .,College of Pharmacy, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 151-742, Republic of Korea.
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, ChungBuk, 363-883, Republic of Korea.
| | - Hyung-Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, ChungBuk, 363-883, Republic of Korea.
| | - Hye-Suk Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 151-742, Republic of Korea.
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 420-743, Republic of Korea.
| |
Collapse
|
93
|
Pan XY, Peng L, Han ZQ, Yin GQ, Song YK, Huang J. Hirudin promotes angiogenesis by modulating the cross-talk between p38 MAPK and ERK in rat ischemic skin flap tissue. Tissue Cell 2015; 47:301-10. [DOI: 10.1016/j.tice.2015.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/24/2015] [Accepted: 04/03/2015] [Indexed: 02/03/2023]
|
94
|
Papageorgiou AP, Heggermont W, Rienks M, Carai P, Langouche L, Verhesen W, De Boer RA, Heymans S. Liver X receptor activation enhances CVB3 viral replication during myocarditis by stimulating lipogenesis. Cardiovasc Res 2015; 107:78-88. [PMID: 25998987 DOI: 10.1093/cvr/cvv157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/13/2015] [Indexed: 12/16/2022] Open
Abstract
AIMS Viral myocarditis (VM) is severe cardiac inflammation that can result in sudden death or congestive heart failure in previously healthy adults, with no effective therapy. Liver X receptor (LXR) agonists have both anti-inflammatory and lipid-lowering properties. This study investigates whether LXR agonist T0901317 may modulate viral replication and cardiac inflammation during VM. METHODS AND RESULTS (i) Adult mice were administered T0901317 or vehicle with the onset of inflammation during CVB3 virus myocarditis or (ii) treated 2 days prior to CVB3 infection. Against what we expected, T0901317 treatment did not alter leucocyte infiltration after CVB3 infection; yet pre-administration with T0901317 resulted in increased mortality upon CVB3 infection, higher cardiac viral presence, and increased cardiomyocyte damage when compared with the vehicle. Furthermore, we show a correlation of fatty acid synthase (FAS) and sterol regulatory element-binding protein 1c (SREBP-1c) with CVB3 viral load in the heart and that T0901317 is able to enhance the cardiac expression of FAS and SREBP-1c. Finally, we show in vitro that T0901317 is able to exaggerate CVB3-mediated damage of Vero cells, whereas inhibitors of FAS and the SREBP-1c reduce the viral presence of CVB3 in neonatal cardiomyocytes. CONCLUSION LXR agonism does not modulate cardiac inflammation, but exacerbates virus-mediated myocardial damage during VM by stimulating lipid biosynthesis and enhancing CVB3 replication.
Collapse
Affiliation(s)
- Anna-Pia Papageorgiou
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Ward Heggermont
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Marieke Rienks
- CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Paolo Carai
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Lies Langouche
- Laboratory of Intensive Care Medicine, KU Leuven, Leuven, Belgium
| | - Wouter Verhesen
- CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Rudolf A De Boer
- University Medical Center, Groningen University, Groningen, The Netherlands
| | - Stephane Heymans
- Centre for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium CArdiovascular Research Institute Maastricht (CARIM), Universiteitssingel 50, 6229 ER Maastricht, The Netherlands ICIN - Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
95
|
Kang YJ, Bang BR, Otsuka M, Otsu K. Tissue-Specific Regulation of p38α-Mediated Inflammation in Con A-Induced Acute Liver Damage. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:4759-4766. [PMID: 25888643 PMCID: PMC4417423 DOI: 10.4049/jimmunol.1402954] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/16/2015] [Indexed: 01/19/2023]
Abstract
Because p38α plays a critical role in inflammation, it has been an attractive target for the development of anti-inflammation therapeutics. However, p38α inhibitors showed side effects, including severe liver toxicity, that often prevailed over the benefits in clinical studies, and the mechanism of toxicity is not clear. In this study, we demonstrate that p38α regulates the inflammatory responses in acute liver inflammation in a tissue-specific manner, and liver toxicity by p38α inhibitors may be a result of the inhibition of protective activity of p38α in the liver. Genetic ablation of p38α in T and NKT cells protected mice from liver injury in Con A-induced liver inflammation, whereas liver-specific deletion of p38α aggravated liver pathology. We found that p38α deficiency in the liver increased the expression of chemokines to recruit more inflammatory cells, indicating that p38α in the liver plays a protective anti-inflammatory role during acute liver inflammation. Therefore, our results suggest that p38α regulates the inflammatory responses in a tissue-specific manner, and that the tissue-specific p38α targeting strategies can be used for the development of an effective anti-inflammation treatment with an improved side-effect profile.
Collapse
Affiliation(s)
- Young Jun Kang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037;
| | - Bo-Ram Bang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Kinya Otsu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan; and Cardiovascular Division, King's College London, London SE5 9NU, United Kingdom
| |
Collapse
|
96
|
Antiproliferative Activity of Hinokitiol, a Tropolone Derivative, Is Mediated via the Inductions of p-JNK and p-PLCγ1 Signaling in PDGF-BB-Stimulated Vascular Smooth Muscle Cells. Molecules 2015; 20:8198-212. [PMID: 25961161 PMCID: PMC6272725 DOI: 10.3390/molecules20058198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/22/2015] [Accepted: 04/28/2015] [Indexed: 12/15/2022] Open
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) is important in the pathogenesis of vascular disorders such as atherosclerosis and restenosis. Hinokitiol, a tropolone derivative found in Chamacyparis taiwanensis, has been found to exhibit anticancer activity in a variety of cancers through inhibition of cell proliferation. In the present study, the possible anti-proliferative effect of hinokitiol was investigated on VSMCs. Our results showed that hinokitiol significantly attenuated the PDGF-BB-stimulated proliferation of VSMCs without cytotoxicity. Hinokitiol suppressed the expression of proliferating cell nuclear antigen (PCNA), a maker for cell cycle arrest, and caused G0/G1 phase arrest in cell cycle progression. To investigate the mechanism underlying the anti-proliferative effect of hinokitiol, we examined the effects of hinokitiol on phosphorylations of Akt, ERK1/2, p38 and JNK1/2. Phospholipase C (PLC)-γ1 phosphorylation, its phosphorylated substrates and p27kip1 expression was also analyzed. Pre-treatment of VSMCs with hinikitiol was found to significantly inhibit the PDGF-BB-induced phosphorylations of JNK1/2 and PLC-γ1, however no effects on Akt, ERK1/2, and p38. The up-regulation of p27kip1 was also observed in hinokitiol-treated VSMCs. Taken together, our results suggest that hinokitiol inhibits PDGF-BB-induced proliferation of VSMCs by inducing cell cycle arrest, suppressing JNK1/2 phosphorylation and PLC-γ1, and stimulating p27kip1 expression. These findings suggest that hinokitiol may be beneficial for the treatment of vascular-related disorders and diseases.
Collapse
|
97
|
Jain M, Singh A, Singh V, Barthwal MK. Involvement of interleukin-1 receptor-associated kinase-1 in vascular smooth muscle cell proliferation and neointimal formation after rat carotid injury. Arterioscler Thromb Vasc Biol 2015; 35:1445-55. [PMID: 25908764 DOI: 10.1161/atvbaha.114.305028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/07/2015] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Reduced frequency of atherosclerotic plaques is observed in interleukin-1 receptor-associated kinase-1 (IRAK1)-deficient mice; however, the underlying mechanism is not clear. Therefore, this study investigate the role of IRAK1 in vascular smooth muscle cell proliferation and neointimal hyperplasia. APPROACH AND RESULTS Stimulation of rat primary vascular smooth muscle cells with fetal bovine serum (10%) or platelet-derived growth factor-BB (20 ng/mL) for 15 minutes to 24 hours induced a time-dependent increase in IRAK1 and extracellular signal-regulated kinase (ERK) activation, proliferating cell nuclear antigen upregulation and p27Kip1 downregulation as assessed by Western blotting. Inhibitors of ERK pathway (U0126, 10 μmol/L), IRAK (IRAK1/4, 3 μmol/L), protein kinase C (PKC; Ro-31-8220, 1 μmol/L), siRNA of toll-like receptor-4 (200 nmol/L), and PKC-ε (200 nmol/L) significantly attenuated these changes. Platelet-derived growth factor induced endogenous IRAK-ERK-PKC-ε association in a toll-like receptor-4 and PKC-ε-dependent manner. A time-dependent increase in IRAK1 and ERK activation was observed after 15 minutes, 30 minutes, 1 hour, 6 hours, 12 hours, and 24 hours of carotid balloon injury in rats. Balloon injury induced endogenous IRAK-ERK-PKC-ε interaction. Perivascular application of IRAK1/4 inhibitor (100 μmol/L), U0126 (100 μmol/L), and IRAK1 siRNA (220 and 360 nmol/L) in pluronic gel abrogated balloon injury-induced ERK phosphorylation, activation, and p27Kip1 downregulation. Hematoxylin and eosin staining and immunohistochemistry of proliferating cell nuclear antigen and smooth muscle actin demonstrated that balloon injury-induced intimal thickening and neointimal vascular smooth muscle cell proliferation were significantly abrogated in the presence of IRAK1/4 inhibitor, IRAK1 siRNA, and U0126. CONCLUSIONS IRAK1 mediates vascular smooth muscle cell proliferation and neointimal hyperplasia by regulating PKC-ε-IRAK1-ERK axis.
Collapse
Affiliation(s)
- Manish Jain
- From the Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ankita Singh
- From the Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vishal Singh
- From the Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Manoj Kumar Barthwal
- From the Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
98
|
Chen YC, Wen ZH, Lee YH, Chen CL, Hung HC, Chen CH, Chen WF, Tsai MC. Dihydroaustrasulfone alcohol inhibits PDGF-induced proliferation and migration of human aortic smooth muscle cells through inhibition of the cell cycle. Mar Drugs 2015; 13:2390-406. [PMID: 25898413 PMCID: PMC4413217 DOI: 10.3390/md13042390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/27/2015] [Accepted: 04/09/2015] [Indexed: 12/26/2022] Open
Abstract
Dihydroaustrasulfone alcohol is the synthetic precursor of austrasulfone, which is a marine natural product, isolated from the Taiwanese soft coral Cladiella australis. Dihydroaustrasulfone alcohol has anti-inflammatory, neuroprotective, antitumor and anti-atherogenic properties. Although dihydroaustrasulfone alcohol has been shown to inhibit neointima formation, its effect on human vascular smooth muscle cells (VSMCs) has not been elucidated. We examined the effects and the mechanisms of action of dihydroaustrasulfone alcohol on proliferation, migration and phenotypic modulation of human aortic smooth muscle cells (HASMCs). Dihydroaustrasulfone alcohol significantly inhibited proliferation, DNA synthesis and migration of HASMCs, without inducing cell death. Dihydroaustrasulfone alcohol also inhibited platelet-derived growth factor (PDGF)-induced expression of cyclin-dependent kinases (CDK) 2, CDK4, cyclin D1 and cyclin E. In addition, dihydroaustrasulfone alcohol inhibited PDGF-induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), whereas it had no effect on the phosphorylation of phosphatidylinositol 3-kinase (PI3K)/(Akt). Moreover, treatment with PD98059, a highly selective ERK inhibitor, blocked PDGF-induced upregulation of cyclin D1 and cyclin E and downregulation of p27kip1. Furthermore, dihydroaustrasulfone alcohol also inhibits VSMC synthetic phenotype formation induced by PDGF. For in vivo studies, dihydroaustrasulfone alcohol decreased smooth muscle cell proliferation in a rat model of restenosis induced by balloon injury. Immunohistochemical staining showed that dihydroaustrasulfone alcohol noticeably decreased the expression of proliferating cell nuclear antigen (PCNA) and altered VSMC phenotype from a synthetic to contractile state. Our findings provide important insights into the mechanisms underlying the vasoprotective actions of dihydroaustrasulfone alcohol and suggest that it may be a useful therapeutic agent for the treatment of vascular occlusive disease.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Aorta/cytology
- Butanones/administration & dosage
- Butanones/pharmacology
- Butanones/therapeutic use
- Cardiovascular Agents/administration & dosage
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Carotid Artery Injuries/drug therapy
- Carotid Artery Injuries/immunology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Carotid Artery, Common/drug effects
- Carotid Artery, Common/immunology
- Carotid Artery, Common/metabolism
- Carotid Artery, Common/pathology
- Cell Cycle/drug effects
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Female
- Gene Expression Regulation/drug effects
- Humans
- Injections, Intraperitoneal
- MAP Kinase Signaling System/drug effects
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/metabolism
- Rats, Sprague-Dawley
- Sulfones/administration & dosage
- Sulfones/pharmacology
- Sulfones/therapeutic use
Collapse
Affiliation(s)
- Yao-Chang Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Lienhai Road, Kaohsiung 804, Taiwan.
- Department of Biomedical Engineering, National Defense Medical Center, Sec. 6, Minquan E. Road, Taipei 11490, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Lienhai Road, Kaohsiung 804, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan.
| | - Yen-Hsien Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei 11042, Taiwan.
| | - Chu-Lun Chen
- Department of Physiology and Biophysics; Graduate Institute of Physiology, National Defense Medical Center, Sec. 6, Minquan E. Road, Taipei 11490, Taiwan.
| | - Han-Chun Hung
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan.
| | - Chun-Hong Chen
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan.
| | - Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| | - Min-Chien Tsai
- Department of Physiology and Biophysics; Graduate Institute of Physiology, National Defense Medical Center, Sec. 6, Minquan E. Road, Taipei 11490, Taiwan.
| |
Collapse
|
99
|
Chen S, Liu B, Kong D, Li S, Li C, Wang H, Sun Y. Atorvastatin calcium inhibits phenotypic modulation of PDGF-BB-induced VSMCs via down-regulation the Akt signaling pathway. PLoS One 2015; 10:e0122577. [PMID: 25874930 PMCID: PMC4398430 DOI: 10.1371/journal.pone.0122577] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/16/2015] [Indexed: 11/30/2022] Open
Abstract
Plasticity of vascular smooth muscle cells (VSMCs) plays a central role in the onset and progression of proliferative vascular diseases. In adult tissue, VSMCs exist in a physiological contractile-quiescent phenotype, which is defined by lack of the ability of proliferation and migration, while high expression of contractile marker proteins. After injury to the vessel, VSMC shifts from a contractile phenotype to a pathological synthetic phenotype, associated with increased proliferation, migration and matrix secretion. It has been demonstrated that PDGF-BB is a critical mediator of VSMCs phenotypic switch. Atorvastatin calcium, a selective inhibitor of 3-hydroxy-3-methyl-glutaryl l coenzyme A (HMG-CoA) reductase, exhibits various protective effects against VSMCs. In this study, we investigated the effects of atorvastatin calcium on phenotype modulation of PDGF-BB-induced VSMCs and the related intracellular signal transduction pathways. Treatment of VSMCs with atorvastatin calcium showed dose-dependent inhibition of PDGF-BB-induced proliferation. Atorvastatin calcium co-treatment inhibited the phenotype modulation and cytoskeleton rearrangements and improved the expression of contractile phenotype marker proteins such as α-SM actin, SM22α and calponin in comparison with PDGF-BB alone stimulated VSMCs. Although Akt phosphorylation was strongly elicited by PDGF-BB, Akt activation was attenuated when PDGF-BB was co-administrated with atorvastatin calcium. In conclusion, atorvastatin calcium inhibits phenotype modulation of PDGF-BB-induced VSMCs and activation of the Akt signaling pathway, indicating that Akt might play a vital role in the modulation of phenotype.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Baoqin Liu
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Dehui Kong
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Si Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Chao Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Huaqin Wang
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang Liaoning, China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
- * E-mail:
| |
Collapse
|
100
|
Duggirala A, Delogu F, Angelini TG, Smith T, Caputo M, Rajakaruna C, Emanueli C. Non coding RNAs in aortic aneurysmal disease. Front Genet 2015; 6:125. [PMID: 25883602 PMCID: PMC4381652 DOI: 10.3389/fgene.2015.00125] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/16/2015] [Indexed: 01/21/2023] Open
Abstract
An aneurysm is a local dilatation of a vessel wall which is >50% its original diameter. Within the spectrum of cardiovascular diseases, aortic aneurysms are among the most challenging to treat. Most patients present acutely after aneurysm rupture or dissection from a previous asymptomatic condition and are managed by open surgical or endovascular repair. In addition, patients may harbor concurrent disease contraindicating surgical intervention. Collectively, these factors have driven the search for alternative methods of identifying, monitoring and treating aortic aneurisms using less invasive approaches. Non-coding RNA (ncRNAs) are emerging as new fundamental regulators of gene expression. The small microRNAs have opened the field of ncRNAs capturing the attention of basic and clinical scientists for their potential to become new therapeutic targets and clinical biomarkers for aortic aneurysm. More recently, long ncRNAs (lncRNAs) have started to be actively investigated, leading to first exciting reports, which further suggest their important and yet largely unexplored contribution to vascular physiology and disease. This review introduces the different ncRNA types and focus at ncRNA roles in aorta aneurysms. We discuss the potential of therapeutic interventions targeting ncRNAs and we describe the research models allowing for mechanistic studies and clinical translation attempts for controlling aneurysm progression. Furthermore, we discuss the potential role of microRNAs and lncRNAs as clinical biomarkers.
Collapse
Affiliation(s)
- Aparna Duggirala
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol Bristol, UK
| | - Francesca Delogu
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol Bristol, UK
| | | | - Tanya Smith
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol Bristol, UK
| | - Massimo Caputo
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol Bristol, UK ; Rush Centre for Congenital and Structural Heart Disease, Rush University Medical Centre Chicago, IL, USA
| | - Cha Rajakaruna
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol Bristol, UK
| | - Costanza Emanueli
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol Bristol, UK
| |
Collapse
|