551
|
Findeisen HM, Gizard F, Zhao Y, Qing H, Heywood EB, Jones KL, Cohn D, Bruemmer D. Epigenetic regulation of vascular smooth muscle cell proliferation and neointima formation by histone deacetylase inhibition. Arterioscler Thromb Vasc Biol 2011; 31:851-60. [PMID: 21233448 DOI: 10.1161/atvbaha.110.221952] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Proliferation of smooth muscle cells (SMC) in response to vascular injury is central to neointimal vascular remodeling. There is accumulating evidence that histone acetylation constitutes a major epigenetic modification for the transcriptional control of proliferative gene expression; however, the physiological role of histone acetylation for proliferative vascular disease remains elusive. METHODS AND RESULTS In the present study, we investigated the role of histone deacetylase (HDAC) inhibition in SMC proliferation and neointimal remodeling. We demonstrate that mitogens induce transcription of HDAC 1, 2, and 3 in SMC. Short interfering RNA-mediated knockdown of either HDAC 1, 2, or 3 and pharmacological inhibition of HDAC prevented mitogen-induced SMC proliferation. The mechanisms underlying this reduction of SMC proliferation by HDAC inhibition involve a growth arrest in the G(1) phase of the cell cycle that is due to an inhibition of retinoblastoma protein phosphorylation. HDAC inhibition resulted in a transcriptional and posttranscriptional regulation of the cyclin-dependent kinase inhibitors p21(Cip1) and p27(Kip). Furthermore, HDAC inhibition repressed mitogen-induced cyclin D1 mRNA expression and cyclin D1 promoter activity. As a result of this differential cell cycle-regulatory gene expression by HDAC inhibition, the retinoblastoma protein retains a transcriptional repression of its downstream target genes required for S phase entry. Finally, we provide evidence that these observations are applicable in vivo by demonstrating that HDAC inhibition decreased neointima formation and expression of cyclin D1 in a murine model of vascular injury. CONCLUSIONS These findings identify HDAC as a critical component of a transcriptional cascade regulating SMC proliferation and suggest that HDAC might play a pivotal role in the development of proliferative vascular diseases, including atherosclerosis and in-stent restenosis.
Collapse
Affiliation(s)
- Hannes M Findeisen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | | | | | | | | | | | | | | |
Collapse
|
552
|
Stawowy P, Kappert K. The molecular biology of furin-like proprotein convertases in vascular remodelling. Methods Mol Biol 2011; 768:191-206. [PMID: 21805243 DOI: 10.1007/978-1-61779-204-5_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration represent key features in atherosclerosis and restenosis. The proprotein convertases (PCs) furin and PC5 are highly expressed in human atheroma and are putatively involved in vascular lesion formation via the activation of precursor proteins, essential for cell proliferation and migration. In vitro assays have identified these PCs to govern cell functions via endoproteolytic cleavage of key substrates, including pro-integrins and pro-matrix metalloproteinases. In vivo gene expression studies of furin/PC5 and their substrates demonstrate their coordinated regulation in animal models of vascular remodelling and in human atherosclerotic lesions. Here we describe in vitro and in vivo models to investigate the function of furin/PC5 in VSMCs and in vascular lesion formation. In conjunction with the development of novel PC inhibitors, this should facilitate the development of new strategies targeting PCs in cardiovascular disease.
Collapse
Affiliation(s)
- Philipp Stawowy
- Department of Medicine/Cardiology, Deutsches Herzzentrum Berlin, D-13353, Berlin, Germany.
| | | |
Collapse
|
553
|
Kang SW, Kim JL, Kwon GT, Lee YJ, Park JHY, Lim SS, Kang YH. Sensitive Fern (Onoclea sensibilis) Extract Suppresses Proliferation and Migration of Vascular Smooth Muscle Cells Inflamed by Neighboring Macrophages. Biol Pharm Bull 2011; 34:1717-23. [DOI: 10.1248/bpb.34.1717] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Jung-Lye Kim
- Department of Food and Nutrition, Hallym University
| | | | | | | | | | | |
Collapse
|
554
|
Huang J, Zhang J, Pathak A, Li J, Stouffer GA. Perivascular delivery of blebbistatin reduces neointimal hyperplasia after carotid injury in the mouse. J Pharmacol Exp Ther 2011; 336:116-26. [PMID: 20956482 PMCID: PMC3014304 DOI: 10.1124/jpet.110.174615] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 10/15/2010] [Indexed: 11/22/2022] Open
Abstract
Proliferation and migration of smooth muscle cells (SMC) require myosin II activity; thus, we examined whether blebbistatin, a cell-permeable selective inhibitor of myosin II ATP activity, would impair neointimal hyperplasia after vascular injury. Delivery of blebbistatin via a perivascular polymer cuff reduced neointimal formation by 73% and luminal obstruction by 75% after carotid denudation injury in C57BL/6 mice. Blebbistatin treatment was also associated with a reduction in cell density within the neointima; total number of cells (76 ± 7 to 27 ± 3 cells/high-powered field) and actin-positive cells (64 ± 4 to 24 ± 2 cells/high-powered field) in the neointima were reduced in blebbistatin-treated mice compared with vehicle-treated mice. In a model of vascular injury with an intact endothelium, implantation of a blebbistatin-secreting cuff after carotid ligation in FVB/N mice was associated with a 61% decrease in neointimal area and a significant decrease in luminal obstruction (88 ± 4% in vehicle-treated mice versus 36 ± 4% in blebbistatin-treated mice; p < 0.0001). In cultured rat aortic SMC, blebbistatin disrupted cellular morphology and actin cytoskeleton structure, and these effects were rapid and completely reversible. Blebbistatin had a dose-dependent inhibitory effect on DNA replication and cell proliferative responses to platelet-derived growth factor-BB, angiotensin II, and α-thrombin, migratory responses to serum, and migratory responses after blunt injury. In summary, perivascular delivery of blebbistatin reduced neointimal hyperplasia after carotid injury in the mouse.
Collapse
Affiliation(s)
- Jianhua Huang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | |
Collapse
|
555
|
Li Y, Yu J, Li M, Qu Z, Ruan Q. Mouse mesenchymal stem cells from bone marrow differentiate into smooth muscle cells by induction of plaque-derived smooth muscle cells. Life Sci 2011; 88:130-40. [DOI: 10.1016/j.lfs.2010.10.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 10/15/2010] [Accepted: 10/27/2010] [Indexed: 02/01/2023]
|
556
|
Francis AA, Pierce GN. An integrated approach for the mechanisms responsible for atherosclerotic plaque regression. Exp Clin Cardiol 2011; 16:77-86. [PMID: 22065938 PMCID: PMC3209544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 07/27/2011] [Indexed: 05/31/2023]
Abstract
Atherosclerosis was originally considered to be an ongoing process that was inevitably associated with age. However, plaques are highly dynamic, and are able to progress, stabilize or regress depending on their surrounding milieu. A great deal of research attention has been focused on understanding the involvement of high-density lipoprotein in atherosclerotic plaque regression. However, atherosclerotic plaque regression encompasses a variety of processes that can be grouped into three main areas: removal of lipids and necrotic material; restoration of endothelial function and repair of denuded areas; and cessation of vascular smooth muscle cell proliferation and phenotype reversal. In addition to the role of high-density lipoproteins in lipid removal, resident macrophages and foam cells are able to regain motility and rapidly migrate on milieu improvement, moving both lipids and necrotic material to regional lymph nodes. Neighbouring endothelial cells can proliferate and replace dead and dysfunctional cells. Circulating endothelial progenitor cells can similarly restore vessel function. Finally, abrogation of smooth muscle cell proliferation occurs secondarily to these processes. This information is integrated in the current article to present a comprehensive and clear depiction of plaque regression. This integrated view of regression is essential to optimize the pharmaceutical targeting of the many processes and pathways involved in plaque regression.
Collapse
Affiliation(s)
| | - Grant N Pierce
- Correspondence: Dr Grant N Pierce, Institute of Cardiovascular Sciences, St Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, Manitoba R2H 2A6. Telephone 204-235-3206, fax 204-235-0793, e-mail
| |
Collapse
|
557
|
Demyanets S, Kaun C, Rychli K, Pfaffenberger S, Kastl SP, Hohensinner PJ, Rega G, Katsaros KM, Afonyushkin T, Bochkov VN, Paireder M, Huk I, Maurer G, Huber K, Wojta J. Oncostatin M-enhanced vascular endothelial growth factor expression in human vascular smooth muscle cells involves PI3K-, p38 MAPK-, Erk1/2- and STAT1/STAT3-dependent pathways and is attenuated by interferon-γ. Basic Res Cardiol 2010; 106:217-31. [PMID: 21174212 DOI: 10.1007/s00395-010-0141-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 11/30/2010] [Accepted: 12/06/2010] [Indexed: 12/21/2022]
Abstract
The pleiotropic cytokine oncostatin M (OSM), a member of the glycoprotein (gp)130 ligand family, plays a key role in inflammation and cardiovascular disease. As inflammation precedes and accompanies pathological angiogenesis, we investigated the effect of OSM and other gp130 ligands on vascular endothelial growth factor (VEGF) production in human vascular smooth muscle cells (SMC). Human coronary artery SMC (HCASMC) and human aortic SMC (HASMC) were treated with different gp130 ligands. VEGF protein was determined by ELISA. Specific mRNA was detected by RT-PCR. Western blotting was performed for signal transducers and activators of transcription1 (STAT1), STAT3, Akt and p38 mitogen-activated protein kinase (p38 MAPK). OSM mRNA and VEGF mRNA expression was analyzed in human carotid endaterectomy specimens from 15 patients. OSM increased VEGF production in both HCASMC and HASMC derived from different donors. OSM upregulated VEGF and OSM receptor-specific mRNA in these cells. STAT3 inhibitor WP1066, p38 MAPK inhibitors SB-202190 and BIRB 0796, extracellular signal-regulated kinase1/2 (Erk1/2) inhibitor U0126, and phosphatidylinositol 3-kinase (PI3K) inhibitors LY-294002 and PI-103 reduced OSM-induced VEGF synthesis. We found OSM expression in human atherosclerotic lesions where OSM mRNA correlated with VEGF mRNA expression. Interferon-γ (IFN-γ), but not IL-4 or IL-10, reduced OSM-induced VEGF production in vascular SMC. Our findings that OSM, which is present in human atherosclerotic lesions and correlates with VEGF expression, stimulates production of VEGF by human coronary artery and aortic SMC indicate that OSM could contribute to plaque angiogenesis and destabilization. IFN-γ reduced OSM-induced VEGF production by vascular SMC.
Collapse
Affiliation(s)
- Svitlana Demyanets
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
558
|
Palozza P, Parrone N, Simone RE, Catalano A. Lycopene in atherosclerosis prevention: An integrated scheme of the potential mechanisms of action from cell culture studies. Arch Biochem Biophys 2010; 504:26-33. [DOI: 10.1016/j.abb.2010.06.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/14/2010] [Accepted: 06/27/2010] [Indexed: 12/31/2022]
|
559
|
Hashizume S, Akaike M, Azuma H, Ishikawa K, Yoshida S, Sumitomo-Ueda Y, Yagi S, Ikeda Y, Iwase T, Aihara KI, Abe M, Sata M, Matsumoto T. Activation of peroxisome proliferator-activated receptor α in megakaryocytes reduces platelet-derived growth factor-BB in platelets. J Atheroscler Thromb 2010; 18:138-47. [PMID: 21060209 DOI: 10.5551/jat.5868] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Platelet-derived growth factor (PDGF)-BB plays a crucial role in atherosclerosis and vascular remodeling by promoting the migration and proliferation of vascular smooth muscle cells. The objective of this study was to clarify the pleiotropic effect of peroxisome proliferator-activated receptor α (PPARα) activators on PDGF-BB expression in megakaryocytes and platelets. METHODS AND RESULTS The expression of PPARα in a human erythroleukemia (HEL) cells was clearly detected by reverse transcriptase-PCR and immunofluorescence microscopy. The expression level of PPARα in HEL cells was unchanged regardless of differentiation into megakaryocytic cells by treatment with phorbol 12-myristate 13 acetate (TPA). The TPA-induced expression of PDGF-B mRNA and PDGF-BB protein levels in culture media was significantly decreased by treatment with PPARα activators, Wy14643 and fenofibric acid, in a dose-dependent manner. PDGF-BB expression induced by inflammatory cytokines, including interleukin-1β or interleukin-6, was also significantly suppressed by treatment with PPARα activators. Immunohistochemistry of human bone marrow showed the expression of PPARα in both the nucleus and cytoplasm of megakaryocytes. In addition, PDGF-BB levels in platelets were significantly decreased from 1,800±870 to 1,470±840 pg/10(5) platelets (mean±SD, p<0.05) by treatment with 300 mg fenofibrate once daily for 4 weeks in 13 patients with dyslipidemia. CONCLUSIONS Activation of PPARα in megakaryocytes reduces PDGF-BB expression in platelets. PPARα activators may exert vasculo-protective action through suppression of PDGF-BB production in a megakaryocyte/platelet pathway.
Collapse
Affiliation(s)
- Shunji Hashizume
- Department of Medicine and Bioregulatory Sciences, The University of Tokushima Graduate School of Health Biosciences, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
560
|
Jung HO, Uhm JS, Seo SM, Kim JH, Youn HJ, Baek SH, Chung WS, Seung KB. Angiotensin II-induced smooth muscle cell migration is mediated by LDL receptor-related protein 1 via regulation of matrix metalloproteinase 2 expression. Biochem Biophys Res Commun 2010; 402:577-82. [DOI: 10.1016/j.bbrc.2010.10.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 10/05/2010] [Indexed: 10/19/2022]
|
561
|
Villeneuve LM, Kato M, Reddy MA, Wang M, Lanting L, Natarajan R. Enhanced levels of microRNA-125b in vascular smooth muscle cells of diabetic db/db mice lead to increased inflammatory gene expression by targeting the histone methyltransferase Suv39h1. Diabetes 2010; 59:2904-15. [PMID: 20699419 PMCID: PMC2963550 DOI: 10.2337/db10-0208] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 07/13/2010] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Diabetes remains a major risk factor for vascular complications that seem to persist even after achieving glycemic control, possibly due to "metabolic memory." Using cultured vascular smooth muscle cells (MVSMC) from type 2 diabetic db/db mice, we recently showed that decreased promoter occupancy of the chromatin histone H3 lysine-9 methyltransferase Suv39h1 and the associated repressive epigenetic mark histone H3 lysine-9 trimethylation (H3K9me3) play key roles in sustained inflammatory gene expression. Here we examined the role of microRNAs (miRs) in Suv39h1 regulation and function in MVSMC from diabetic mice. RESEARCH DESIGN AND METHODS We used luciferase assays with Suv39h1 3'untranslated region (UTR) reporter constructs and Western blotting of endogenous protein to verify that miR-125b targets Suv39h1. We examined the effects of Suv39h1 targeting on inflammatory gene expression by quantitative real time polymerase chain reaction (RT-qPCR), and H3K9me3 levels at their promoters by chromatin immunoprecipitation assays. RESULTS We observed significant upregulation of miR-125b with parallel downregulation of Suv39h1 protein (predicted miR-125b target) in MVSMC cultured from diabetic db/db mice relative to control db/+. miR-125b mimics inhibited both Suv39h1 3'UTR luciferase reporter activity and endogenous Suv39h1 protein levels. Conversely, miR-125b inhibitors showed opposite effects. Furthermore, miR-125b mimics increased expression of inflammatory genes, monocyte chemoattractant protein-1, and interleukin-6, and reduced H3K9me3 at their promoters in nondiabetic cells. Interestingly, miR-125b mimics increased monocyte binding to db/+ MVSMC toward that in db/db MVSMC, further imitating the proinflammatory diabetic phenotype. In addition, we found that the increase in miR-125b in db/db VSMC is caused by increased transcription of miR-125b-2. CONCLUSIONS These results demonstrate a novel upstream role for miR-125b in the epigenetic regulation of inflammatory genes in MVSMC of db/db mice through downregulation of Suv39h1.
Collapse
Affiliation(s)
- Louisa M. Villeneuve
- From the Department of Diabetes, Beckman Research Institute of City of Hope, Duarte, California
| | - Mitsuo Kato
- From the Department of Diabetes, Beckman Research Institute of City of Hope, Duarte, California
| | - Marpadga A. Reddy
- From the Department of Diabetes, Beckman Research Institute of City of Hope, Duarte, California
| | - Mei Wang
- From the Department of Diabetes, Beckman Research Institute of City of Hope, Duarte, California
| | - Linda Lanting
- From the Department of Diabetes, Beckman Research Institute of City of Hope, Duarte, California
| | - Rama Natarajan
- From the Department of Diabetes, Beckman Research Institute of City of Hope, Duarte, California
| |
Collapse
|
562
|
Shagdarsuren E, Bidzhekov K, Mause SF, Simsekyilmaz S, Polakowski T, Hawlisch H, Gessner JE, Zernecke A, Weber C. C5a Receptor Targeting in Neointima Formation After Arterial Injury in Atherosclerosis-Prone Mice. Circulation 2010; 122:1026-36. [DOI: 10.1161/circulationaha.110.954370] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Receptor binding of complement C5a leads to proinflammatory activation of many cell types, but the role of receptor-mediated action during arterial remodeling after injury has not been studied. In the present study, we examined the contribution of the C5a receptor (C5aR) to neointima formation in apolipoprotein E–deficient mice employing a C5aR antagonist (C5aRA) and a C5aR-blocking monoclonal antibody.
Methods and Results—
Mice fed an atherogenic diet were subjected to wire-induced endothelial denudation of the carotid artery and treated with C5aRA and anti-C5aR-blocking monoclonal antibody or vehicle control. Compared with controls, neointima formation was significantly reduced in mice receiving C5aRA or anti-C5aR-blocking monoclonal antibody for 1 week but not for 3 weeks, attributable to an increased content of vascular smooth muscle cells, whereas a marked decrease in monocyte and neutrophil content was associated with reduced vascular cell adhesion molecule-1. As assessed by immunohistochemistry, reverse transcription polymerase chain reaction, and flow cytometry, C5aR was expressed in lesional and cultured vascular smooth muscle cells, upregulated by injury or tumor necrosis factor-α, and reduced by C5aRA. Plasma levels and neointimal plasminogen activator inhibitor-1 peaked 1 week after injury and were downregulated in C5aRA-treated mice. In vitro, C5a induced plasminogen activator inhibitor-1 expression in endothelial cells and vascular smooth muscle cells in a C5aRA-dependent manner, possibly accounting for higher vascular smooth muscle cell immigration.
Conclusions—
One-week treatment with C5aRA or anti-C5aR-blocking monoclonal antibody limited neointimal hyperplasia and inflammatory cell content and was associated with reduced vascular cell adhesion molecule-1 expression. However, treatment for 3 weeks failed to reduce but rather stabilized plaques, likely by reducing vascular plasminogen activator inhibitor-1 and increasing vascular smooth muscle cell migration.
Collapse
Affiliation(s)
- Erdenechimeg Shagdarsuren
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - Kiril Bidzhekov
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - Sebastian F. Mause
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - Sakine Simsekyilmaz
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - Thomas Polakowski
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - Heiko Hawlisch
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - J. Engelbert Gessner
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - Alma Zernecke
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| | - Christian Weber
- From the Institute of Molecular Cardiovascular Research (E.S., K.B., S.F.M., S.S., A.Z., C.W.) and the Department of Cardiology (S.F.M.), RWTH Aachen University, Aachen, Germany; Jerini AG, Berlin, Germany (T.P., H.H.); DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany (A.Z.); Laboratory for Molecular Immunology, Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany (J.E.G.); and Cardiovascular Research Institute Maastricht,
| |
Collapse
|
563
|
Cifuentes RA, Cruz-Tapias P, Rojas-Villarraga A, Anaya JM. ZC3H12A (MCPIP1): molecular characteristics and clinical implications. Clin Chim Acta 2010; 411:1862-8. [PMID: 20807520 DOI: 10.1016/j.cca.2010.08.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Revised: 08/22/2010] [Accepted: 08/24/2010] [Indexed: 12/15/2022]
Abstract
BACKGROUND ZC3H12A is a gene whose absence is related to autoimmune disorders and to other phenotypical alterations. METHODS A comprehensive review of the structure, molecular functions and regulation of ZC3H12A gene and its protein MCPIP1 is done in order to understand their clinical implications. RESULTS ZC3H12A, at 1p34.3, has 9860bp, six exons and 61 described SNPs. Eleven are non-synonymous thus leading to changes in MCPIP1, the protein encoded by ZC3H12A. MCPIP1 is induced by MCP-1 and IL-1 whose signals are transduced through the NF-kβ and MAPkinase pathways. This protein acts as an RNAse by degrading chemokine transcripts such as IL-1 as well as its own mRNA and as a transcription factor by reducing the expression of other chemokines induced by NF-kβ such as MCP-1. It also up-regulates genes involved in several differentiation processes and apoptosis. Therefore, ZC3H12A is an equilibrium gatekeeper that not only regulates its own inducers but also controls the regulation by degrading its own mRNA. CONCLUSION Understanding ZC3H12A gives a comprehensive panorama that promises to improve our understanding of processes in which this gene is involved including autoimmune, infectious and cardiovascular diseases.
Collapse
Affiliation(s)
- Ricardo A Cifuentes
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | | | | | | |
Collapse
|
564
|
Subramanian V, Golledge J, Ijaz T, Bruemmer D, Daugherty A. Pioglitazone-induced reductions in atherosclerosis occur via smooth muscle cell-specific interaction with PPAR{gamma}. Circ Res 2010; 107:953-8. [PMID: 20798360 DOI: 10.1161/circresaha.110.219089] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Peroxisome proliferator-activated receptor (PPAR)γ agonists attenuate atherosclerosis and abdominal aortic aneurysms (AAAs). PPARγ, a nuclear receptor, is expressed on many cell types including smooth muscle cells (SMCs). OBJECTIVE To determine whether a PPARγ agonist reduces angiotensin II (Ang II)-induced atherosclerosis and AAAs via interaction with SMC-specific PPARγ. METHODS AND RESULTS Low-density lipoprotein receptor (LDLR)(-/-) mice with SMC-specific PPARγ deficiency were developed using PPARγ floxed (PPARγ(f/f)) and SM22 Cre(+) mice. PPARγ(f/f) littermates were generated that did not express Cre (Cre(0/0)) or were hemizygous for Cre (Cre(+/0)). To assess the contribution of SMC-specific PPARγ in ligand-mediated attenuation of Ang II-induced atherosclerosis and AAAs, both male and female Cre(0/0) and Cre(+/0) mice were fed a fat-enriched diet with or without the PPARγ agonist pioglitazone (Pio) (20 mg/kg per day) for 5 weeks. After 1 week of feeding modified diets, mice were infused with Ang II (1000 ng/kg per minute) for 4 weeks. SMC-specific PPARγ deficiency or Pio administration had no effect on plasma cholesterol concentrations. Pio administration attenuated Ang II-increased systolic blood pressure equivalently in both Cre(0/0) and Cre(+/0) groups. SMC-specific PPARγ deficiency increased atherosclerosis in male mice. Pio administration reduced atherosclerosis in only the Cre(0/0) mice, but not in mice with SMC-specific PPARγ deficiency. SMC-specific PPARγ deficiency or Pio administration had no effect on Ang II-induced AAA development. Pio also did not attenuate Ang II-induced monocyte chemoattractant protein-1 production in PPARγ-deficient SMCs. CONCLUSIONS Pio attenuates Ang II-induced atherosclerosis via the interaction with SMC-specific PPARγ, but has no effect on the development of AAAs.
Collapse
Affiliation(s)
- Venkateswaran Subramanian
- Saha Cardiovascular Research Center, BBSRB, Room B243, University of Kentucky, Lexington, KY 40536-0509, USA
| | | | | | | | | |
Collapse
|
565
|
Shai SY, Sukhanov S, Higashi Y, Vaughn C, Kelly J, Delafontaine P. Smooth muscle cell-specific insulin-like growth factor-1 overexpression in Apoe-/- mice does not alter atherosclerotic plaque burden but increases features of plaque stability. Arterioscler Thromb Vasc Biol 2010; 30:1916-24. [PMID: 20671230 DOI: 10.1161/atvbaha.110.210831] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Growth factors may play a permissive role in atherosclerosis initiation and progression, in part via their promotion of vascular smooth muscle cell (VSMC) accumulation in plaques. However, unstable human plaques often have a relative paucity of VSMC, which has been suggested to contribute to plaque rupture and erosion and to clinical events. Insulin-like growth factor-1 (IGF-1) is an endocrine and autocrine/paracrine growth factor that is a mitogen for VSMC, but when infused into Apoe(-/-) mice it paradoxically reduces atherosclerosis burden. METHODS AND RESULTS To determine the effect of stimulation of VSMC growth on atherosclerotic plaque development and to understand mechanisms of IGF-1's atheroprotective effect, we assessed atherosclerotic plaques in mice overexpressing IGF-1 in smooth muscle cells (SMC) under the control of the α-smooth muscle actin promoter, after backcrossing to the Apoe(-/-) background (SMP8/Apoe(-/-)). Compared with Apoe(-/-) mice, these SMP8/Apoe(-/-) mice developed a comparable plaque burden after 12 weeks on a Western diet, suggesting that the ability of increased circulating IGF-1 to reduce plaque burden was mediated in large part via non-SMC target cells. However, advanced plaques in SMP8/Apoe(-/-) mice displayed several features of plaque stability, including increased fibrous cap area, α-smooth muscle actin-positive SMC and collagen content, and reduced necrotic cores. CONCLUSIONS These findings indicate that stimulation of VSMC IGF-1 signaling does not alter total atherosclerotic plaque burden and may improve atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Shaw-Yung Shai
- Tulane University Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, La 70112, USA
| | | | | | | | | | | |
Collapse
|
566
|
Mause SF, Ritzel E, Liehn EA, Hristov M, Bidzhekov K, Müller-Newen G, Soehnlein O, Weber C. Platelet microparticles enhance the vasoregenerative potential of angiogenic early outgrowth cells after vascular injury. Circulation 2010; 122:495-506. [PMID: 20644015 DOI: 10.1161/circulationaha.109.909473] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Angiogenic early outgrowth cells (EOCs) have been reported to contribute to endothelial regeneration and to limit neointima formation after vascular injury. Vascular pathologies comprise platelet activation and concomitant generation of platelet microparticles (PMPs). We hypothesized that PMPs may interact with EOCs in the context of vascular injury and modulate their regenerative potential. METHODS AND RESULTS Using flow cytometry, confocal microscopy, and scanning electron microscopy, we demonstrated the binding of thrombin/collagen-induced PMPs to EOCs with subsequent membrane assimilation and incorporation. This interaction promoted phenotypic alterations of EOCs with increased expression of endothelial cell markers and transfer of the chemokine receptor CXCR4 to EOCs with enhanced responsiveness to its ligand CXCL12/SDF-1alpha. In addition, PMPs augmented the adhesion of EOCs to extracellular matrix components and to the injured vessel wall and accelerated cytoskeletal reorganization and migration of EOCs. PMPs induced changes in the EOC secretome toward a more proangiogenic profile and amplified the EOC-mediated induction of proliferation, migration, and capillary tube formation by mature endothelial cells. Compared with untreated EOCs, the injection of PMP-treated EOCs resulted in accelerated reendothelialization after arterial denudation injury in athymic nude mice, whereas the EOC-mediated reduction of neointima formation remained unchanged. CONCLUSIONS Our data provide evidence that PMPs can boost the potential of EOCs to restore endothelial integrity after vascular injury. Major mechanisms involve the enhancement of EOC recruitment, migration, differentiation, and release of proangiogenic factors.
Collapse
Affiliation(s)
- Sebastian F Mause
- Medical Faculty, Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
567
|
Kim MJ, Kwak JH, Baek SH, Yeo HY, Song JH, Cho BJ, Kim OY. Diallyl Sulfides (DAS) and Diallyl Disulfides (DADS) Exhibit a Suppressive Effect on the Proliferation and Migration of Vascular Smooth Muscle. Prev Nutr Food Sci 2010. [DOI: 10.3746/jfn.2010.15.2.137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
568
|
Lamon BD, Upmacis RK, Deeb RS, Koyuncu H, Hajjar DP. Inducible nitric oxide synthase gene deletion exaggerates MAPK-mediated cyclooxygenase-2 induction by inflammatory stimuli. Am J Physiol Heart Circ Physiol 2010; 299:H613-23. [PMID: 20543082 DOI: 10.1152/ajpheart.00144.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclooxygenase (COX)-2 and inducible nitric oxide (NO) synthase (iNOS) are responsive to a wide array of inflammatory stimuli, have been localized to vascular smooth muscle cells (SMCs), and are intimately linked to the progression of vascular disease, including atherosclerotic lesion formation. We and others have shown that the production and subsequent impact of COX products appear to be correlative with the status of NO synthesis. This study examined the impact of inflammation-driven NO production on COX-2 expression in SMCs. Concurrent stimulation of quiescent rat aortic SMCs with lipopolysaccharide (LPS) and interferon (IFN)-gamma increased COX-2, iNOS, and nitrite production. Pharmacological inhibition of NO synthase (N(G)-monomethyl-l-arginine) concentration- and time-dependently magnified LPS + IFN-gamma-mediated COX-2 mRNA and protein induction in a cGMP-independent manner. COX-2 induction was associated with activation of the ERK, p38, and JNK mitogen-activated protein kinase (MAPK) pathways. Interestingly, NO synthase inhibition enhanced ERK, p38, and to a lesser extent JNK phosphorylation but suppressed MAPK phosphatase (MKP)-1 induction in response to LPS + IFN-gamma. Similarly, the exposure of SMCs from iNOS(-/-) mice to LPS + IFN-gamma produced an enhancement of COX-2 induction, p38, and JNK phosphorylation and an attenuated upregulation of MKP-1 versus their wild-type counterparts. Taken together, our data indicate that NO, in part derived from iNOS, negatively regulates the immediate early induction of COX-2 in response to inflammatory stimuli.
Collapse
Affiliation(s)
- Brian D Lamon
- Department of Pathology and Laboratory Medicine, Center of Vascular Biology, Weill Cornell Medical College of CornellUniversity, New York, New York 10065, USA.
| | | | | | | | | |
Collapse
|
569
|
Kaimoto T, Yasuda O, Ohishi M, Mogi M, Takemura Y, Suhara T, Ogihara T, Fukuo K, Rakugi H. Nifedipine inhibits vascular smooth muscle cell dedifferentiation via downregulation of Akt signaling. Hypertension 2010; 56:247-52. [PMID: 20530298 DOI: 10.1161/hypertensionaha.110.149781] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Calcium is an essential signaling molecule that controls vascular smooth muscle cell (VSMC) contraction, proliferation, and differentiation. Here, we show that the calcium antagonist nifedipine inhibits VSMC dedifferentiation in vitro and in vivo. Differentiated VSMCs cultured on laminin-coated dishes were transferred to laminin-free dishes to induce dedifferentiation. Induction of dedifferentiation resulted in the upregulation of nonmuscle myosin heavy chain expression, a marker of dedifferentiation, and the downregulation of smooth muscle myosin heavy chain expression, a marker of differentiation. Nifedipine significantly inhibited both the induction of these phenotypic changes and upregulation of Akt signaling in these cells. Administration of nifedipine at a low concentration that did not affect blood pressure could inhibit the increase in nonmuscle myosin heavy chain expression and decrease in smooth muscle myosin heavy chain expression in a rat balloon-injury model. Furthermore, nifedipine suppressed neointimal hyperplasia and upregulation of Akt signaling. However, phospho-Akt expression was not suppressed in the regenerating arterial endothelium of the nifedipine-treated rats. The inhibitory effect of the downregulation of Akt signaling by dominant-negative Akt on the induction of VSMC dedifferentiation in the intima was identical to that of nifedipine. In contrast, upregulation of Akt signaling by transfection of the cells with a constitutively active Akt reversed the nifedipine-induced inhibition of VSMC dedifferentiation. In conclusion, nifedipine inhibits VSMC dedifferentiation by suppressing Akt signaling, thereby preventing neointimal thickening.
Collapse
Affiliation(s)
- Taeko Kaimoto
- Department of Cardiovascular Clinical and Translational Research, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
570
|
Role of cellular bioenergetics in smooth muscle cell proliferation induced by platelet-derived growth factor. Biochem J 2010; 428:255-67. [PMID: 20331438 DOI: 10.1042/bj20100090] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Abnormal smooth muscle cell proliferation is a hallmark of vascular disease. Although growth factors are known to contribute to cell hyperplasia, the changes in metabolism associated with this response, particularly mitochondrial respiration, remain unclear. Given the increased energy requirements for proliferation, we hypothesized that PDGF (platelet-derived growth factor) would stimulate glycolysis and mitochondrial respiration and that this elevated bioenergetic capacity is required for smooth muscle cell hyperplasia. To test this hypothesis, cell proliferation, glycolytic flux and mitochondrial oxygen consumption were measured after treatment of primary rat aortic VSMCs (vascular smooth muscle cells) with PDGF. PDGF increased basal and maximal rates of glycolytic flux and mitochondrial oxygen consumption; enhancement of these bioenergetic pathways led to a substantial increase in the mitochondrial reserve capacity. Interventions with the PI3K (phosphoinositide 3-kinase) inhibitor LY-294002 or the glycolysis inhibitor 2-deoxy-D-glucose abrogated PDGF-stimulated proliferation and prevented augmentation of glycolysis and mitochondrial reserve capacity. Similarly, when L-glucose was substituted for D-glucose, PDGF-dependent proliferation was abolished, as were changes in glycolysis and mitochondrial respiration. Interestingly, LDH (lactate dehydrogenase) protein levels and activity were significantly increased after PDGF treatment. Moreover, substitution of L-lactate for D-glucose was sufficient to increase mitochondrial reserve capacity and cell proliferation after treatment with PDGF; these effects were inhibited by the LDH inhibitor oxamate. These results suggest that glycolysis, by providing substrates that enhance the mitochondrial reserve capacity, plays an essential role in PDGF-induced cell proliferation, underscoring the integrated metabolic response required for proliferation of VSMCs in the diseased vasculature.
Collapse
|
571
|
Isenovic ER, Kedees MH, Haidara MA, Trpkovic A, Mikhailidis DP, Marche P. Involvement of ERK1/2 kinase in insulin-and thrombin-stimulated vascular smooth muscle cell proliferation. Angiology 2010; 61:357-364. [PMID: 20304866 DOI: 10.1177/0003319709358693] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
It is well recognized that the proliferation of vascular smooth muscle cells (VSMCs) is a key event in the pathogenesis of various vascular diseases, including atherosclerosis and hypertension. We have previously shown that among extracellular signal-regulated protein kinases (ERKs), the 42- and 44-kDa isoforms (ERK1/2) participate in the cellular mitogenic machinery triggered by several VSMCs activators, including insulin (INS) and thrombin (Thr). However, understanding of the intracellular signal transduction pathways involved is incomplete. This review considers the recent findings in INS and Thr signaling mechanisms that modulate the proliferation of VSMCs with particular emphasis on the ERK1/2 signaling pathway, an important mediator of VSMCs hypertrophy and vascular disease. Moreover, because the ERK1/2 pathway have been acknowledged as an important mediator of VSMCs hypertrophy, ERK1/2 is identified as a key target for novel therapeutic interventions to minimize irreversible tissue damage associated with hypertension and atherosclerosis.
Collapse
Affiliation(s)
- Esma R Isenovic
- Vinca Institute of Nuclear Sciences, University of Belgrade, Laboratory for Molecular Genetics and Radiobiology, Belgrade, Serbia.
| | | | | | | | | | | |
Collapse
|
572
|
Cardiovascular disease in kidney transplant recipients: the prognostic value of inflammatory cytokine genotypes. Transplantation 2010; 89:1001-8. [PMID: 20061995 DOI: 10.1097/tp.0b013e3181ce243f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) represents the main cause of morbidity and mortality after renal transplantation. In view of the modern paradigm of atherosclerosis as an inflammatory disease, this study investigated the impact of inflammatory cytokine polymorphisms on posttransplant CVD. METHODS The association between cytokine polymorphisms and CVD was assessed in a case-control study to identify the differences in genotype distributions between kidney allografts with or without posttransplant CVD. To validate our results in two independent groups, we divided a cohort of 798 renal transplant recipients according to geographic area: an evaluation cohort of 478 patients from Emilia-Romagna and a validation cohort of 320 patients from the rest of Italy. Tumor necrosis factor (TNF)-alpha, transforming growth factor-beta1, interleukin (IL)-10, IL-6, interferon-gamma, and IL-8 polymorphisms were analyzed, and thereafter, the cytokine production genotype was assigned. RESULTS In the evaluation cohort, the patients in the CVD and no-CVD groups differed significantly in TNF-alpha and IL-10 genotype frequencies. Using multivariate analyses to test the association with CVD, the TNF-alpha high-producer genotype was associated with a significantly increased cardiovascular risk (odds ratio [OR]=4.41, 95% confidence interval (CI)=2.53-7.67). Conversely, the IL-10 high-producer genotype resulted protective against CVD (OR=0.07, 95% CI=0.02-0.29). These findings were confirmed in the validation cohort where the carriers of the TNF-alpha high-producer genotype proved to be at 2.45-fold increased cardiovascular risk (OR=2.45, 95% CI=1.29-4.63), whereas the IL-10 high-producer genotype was associated with a 0.08-fold reduced risk (OR=0.08, 95% CI=0.02-0.36). CONCLUSIONS This work suggests a prognostic value of TNF-alpha and IL-10 genotypes, which might represent cardiovascular risk markers in renal transplant.
Collapse
|
573
|
Eder K, Ringseis R. Metabolism and actions of conjugated linoleic acids on atherosclerosis-related events in vascular endothelial cells and smooth muscle cells. Mol Nutr Food Res 2010; 54:17-36. [PMID: 19760681 DOI: 10.1002/mnfr.200900042] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Conjugated linoleic acids (CLAs) are biologically highly active lipid compounds that have attracted great scientific interest due to their ability to cause either inhibition of atherosclerotic plaque development or even regression of pre-established atherosclerotic plaques in mice, hamsters and rabbits. The underlying mechanisms of action, however, are only poorly understood. Since cell culture experiments are appropriate to gain insight into the mechanisms of action of a compound, the present review summarizes data from cell culture studies about the metabolism and the actions of CLAs on atherosclerosis-related events in endothelial cells (ECs) and smooth muscle cells (SMCs), which are important cells contributing to atherosclerotic lesion development. Based on these studies, it can be concluded that CLAs exert several beneficial actions including inhibition of inflammatory and vasoactive mediator release from ECs and SMCs, which may help explain the anti-atherogenic effect of CLAs observed in vivo. The observation that significant levels of CLA metabolites, which have been reported to have significant biological activities, are well detectable in ECs and SMCs indicates that the anti-atherogenic effects observed with CLAs are presumably mediated not only by CLAs themselves but also by their metabolites.
Collapse
Affiliation(s)
- Klaus Eder
- Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising, Germany
| | | |
Collapse
|
574
|
Abstract
The endothelium is critical for the maintenance of a proper vessel function. Disturbances of endothelial function, called endothelial dysfunction, have serious implications, and lead to the development of atherosclerosis. It is well established that the risk for atherosclerosis development is influenced by nutritional factors such as the intake of certain fatty acids. Due to the fundamental role of the endothelium for atherosclerosis development, it is, therefore, likely that fatty acids directly influence the function of endothelial cells. The present review aims to explain the divergent effects of different types of fatty acids on cardiovascular disease risk by summarizing in vitro-data on the effects of fatty acids on (1) important signalling pathways involved in the modulation of endothelial cell function, and (2) endothelial cell functional properties, namely vasoactive mediator release and mononuclear cell recruitment, both of which are typically dysregulated during endothelial dysfunction.
Collapse
Affiliation(s)
- Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Giessen, Giessen, Germany
| | | |
Collapse
|
575
|
Abstract
BACKGROUND AND AIM Pioglitazone has diverse multiple effects on metabolic and inflammatory processes that have the potential to influence cardiovascular disease pathophysiology at various points in the disease process, including atherogenesis, plaque inflammation, plaque rupture, haemostatic disturbances and microangiopathy. RESULTS Linking the many direct and indirect effects on the vasculature to the reduction in key macrovascular outcomes reported with pioglitazone in patients with type 2 diabetes presents a considerable challenge. However, recent large-scale clinical cardiovascular imaging studies are beginning to provide some mechanistic insights, including a potentially important role for improvements in high-density lipoprotein cholesterol with pioglitazone. In addition to a role in prevention, animal studies also suggest that pioglitazone may minimize damage and improve recovery during and after ischaemic cardio- and cerebrovascular events. DESIGN AND METHODS In this review, we consider potential cardiovascular protective mechanisms of pioglitazone by linking preclinical data and clinical cardiovascular outcomes guided by insights from recent imaging studies. CONCLUSION Pioglitazone may influence CVD pathophysiology at multiple points in the disease process, including atherogenesis, plaque inflammation, plaque rupture and haemostatic disturbances (i.e. thrombus/embolism formation), as well as microangiopathy.
Collapse
Affiliation(s)
- E Erdmann
- Department of Medicine, Heart Center, University of Cologne, Cologne, Germany.
| | | |
Collapse
|
576
|
Ma KL, Varghese Z, Ku Y, Powis SH, Chen Y, Moorhead JF, Ruan XZ. Sirolimus inhibits endogenous cholesterol synthesis induced by inflammatory stress in human vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2010; 298:H1646-51. [PMID: 20348217 DOI: 10.1152/ajpheart.00492.2009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammatory stress accelerates the progression of atherosclerosis. Sirolimus, a new immunosuppressive agent, has been shown to have pleiotropic antiatherosclerotic effects. In this study we hypothesized that sirolimus inhibits 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR)-mediated cholesterol synthesis in human vascular smooth muscle cells (VSMCs) under inflammatory stress. Using radioactive assay, we demonstrated that sirolimus inhibited the increase of interleukin-1beta (IL-1beta)-induced cholesterol synthesis in VSMCs. Further studies showed that sirolimus inhibited both the HMGR gene and protein expression in VSMCs treated with or without IL-1beta. These effects were mediated by inhibiting the gene expression of sterol regulatory element-binding protein-2 (SREBP-2) and SREBP-2 cleavage-activating protein (SCAP) as checked by real-time PCR, Western blot analysis, and confocal microscopy for the observation of decreased protein translocation of the SCAP/SREBP-2 complex from the endoplasmic reticulum (ER) to the Golgi. Insulin-induced gene-1 (Insig-1) is a key ER protein controlling the feedback regulation of HMGR at transcriptional and posttranscriptional levels. We demonstrated that sirolimus increased Insig-1 expression which may bind to the SCAP, preventing the exit of SCAP-SREBP complexes from the ER. The increased Insig-1 also accelerated HMGR protein degradation in VSMCs as shown by pulse-chase analysis. In conclusion, sirolimus inhibits cholesterol synthesis induced by inflammatory stress through the downregulation of HMGR expression and the acceleration of HMGR protein degradation. These findings may improve our understanding of the molecular mechanisms of the antiatherosclerosis properties of sirolimus.
Collapse
Affiliation(s)
- Kun L Ma
- Centre for Nephrology, Univ. College London Medical School, Royal Free campus, Rowland Hill St., London, NW3 2PF, UK
| | | | | | | | | | | | | |
Collapse
|
577
|
Matsumoto T, Kobayashi T, Kamata K. Diabetic conditions act as matchmaker for monocytes and vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2010; 298:H731-3. [DOI: 10.1152/ajpheart.01157.2009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Katsuo Kamata
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
578
|
Rombach SM, Twickler TB, Aerts JMFG, Linthorst GE, Wijburg FA, Hollak CEM. Vasculopathy in patients with Fabry disease: current controversies and research directions. Mol Genet Metab 2010; 99:99-108. [PMID: 19900828 DOI: 10.1016/j.ymgme.2009.10.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 10/09/2009] [Indexed: 11/28/2022]
Abstract
Fabry disease is an X-linked lysosomal storage disorder due to deficiency of the enzyme alpha-galactosidase A. The principal clinical manifestations of Fabry disease consist of cardiovascular complications including cerebrovascular, renal and cardiac disease but the pathophysiology of this specific vasculopathy is unclear. With the development of targeted treatment for Fabry disease, i.e. enzyme replacement therapy, it has become apparent that the removal of stored glycosphingolipid from the endothelial cells does not prevent progression of vascular disease in many patients. The aim of this study is to review the current available literature on vascular function tests, imaging and pathology studies and propose a hypothesis on the evolution of arterial complications in Fabry disease. Clearly, although premature atherosclerosis is suggested to occur, most studies describe absence of characteristic plaque formation. Smooth muscle cell hypertrophy, is probably the earliest feature of a complex vasculopathy, as in females and atypical cardiac variants, who have residual enzyme activity, no endothelial storage of significance is found. Subsequently, processes occur as observed in neo intima formation however with formation of more fibrotic structures. In the presence of a hyperdynamic circulation in combination with a less compliant vascular wall, it is hypothesized that upregulation of local renin angiotensine systems may occur. Angiotensin II is known to increase adhesion molecules, cytokines and chemokines and exerts a pro-inflammatory effect on leucocytes, endothelial cells and vascular smooth muscle cells. This enhances release of pro-thrombotic factors and opposes actions mediated through angiotensin 2 (AT2) receptor, including the release of nitric oxide (NO). A combination of reduced vascular compliance and activation of pro-thrombotic factors can lead to vascular complications in Fabry disease.
Collapse
Affiliation(s)
- S M Rombach
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
579
|
Gerli R, Vaudo G, Bocci EB, Schillaci G, Alunno A, Luccioli F, Hijazi R, Mannarino E, Shoenfeld Y. Functional impairment of the arterial wall in primary Sjögren's syndrome: Combined action of immunologic and inflammatory factors. Arthritis Care Res (Hoboken) 2010; 62:712-8. [DOI: 10.1002/acr.20117] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
580
|
Young EWK, Simmons CA. Macro- and microscale fluid flow systems for endothelial cell biology. LAB ON A CHIP 2010; 10:143-60. [PMID: 20066241 DOI: 10.1039/b913390a] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Recent advances in microfluidics have brought forth new tools for studying flow-induced effects on mammalian cells, with important applications in cardiovascular, bone and cancer biology. The plethora of microscale systems developed to date demonstrate the flexibility of microfluidic designs, and showcase advantages of the microscale that are simply not available at the macroscale. However, the majority of these systems will likely not achieve widespread use in the biological laboratory due to their complexity and lack of user-friendliness. To gain widespread acceptance in the biological research community, microfluidics engineers must understand the needs of cell biologists, while biologists must be made aware of available technology. This review provides a critical evaluation of cell culture flow (CCF) systems used to study the effects of mechanical forces on endothelial cells (ECs) in vitro. To help understand the need for various designs of CCF systems, we first briefly summarize main properties of ECs and their native environments. Basic principles of various macro- and microscale systems are described and evaluated. New opportunities are uncovered for developing technologies that have potential to both improve efficiency of experimentation as well as answer important biological questions that otherwise cannot be tackled with existing systems. Finally, we discuss some of the unresolved issues related to microfluidic cell culture, suggest possible avenues of investigation that could resolve these issues, and provide an outlook for the future of microfluidics in biological research.
Collapse
Affiliation(s)
- Edmond W K Young
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA.
| | | |
Collapse
|
581
|
Stouffer GA, Pathak A, Rojas M. Unilateral renal artery stenosis causes a chronic vascular inflammatory response in ApoE-/- mice. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2010; 121:252-266. [PMID: 20697566 PMCID: PMC2917126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
UNLABELLED Unilateral renal artery ligation (RAL) causes a short-term vascular inflammatory reaction in ApoE-/- mice, but whether this persists is unknown. Partial constriction of the right renal artery resulted in a mild, sustained increase in systolic blood pressure over the 90 days of the study, whereas there was no change in blood pressure in mice that underwent sham-surgery. Atheroma was present in the abdominal aorta, with pronounced staining for macrophages (39% +/- 9% of area of atheroma) and monocyte chemoattractant-1 (MCP-1; 40%+/-15%) in mice with unilateral RAL. Atheroma was also present in the carotid arteries, with pronounced staining for macrophages (56%+/-15%) and MCP-1 (53%+/-21%). Non-muscle myosin-A, a marker of smooth muscle cell dedifferentiation, was observed in atheroma within the abdominal aorta and carotid arteries in mice that underwent unilateral RAL. There were no atheroma and minimal staining for macrophages, MCP-1 or non-muscle myosin A in carotid arteries or abdominal aorta of mice that underwent sham surgery. CONCLUSIONS Chronic unilateral partial RAL results in the formation of atheroma in the aorta and carotid arteries of ApoE-/- mice that is characterized by chronic inflammation and dedifferentiation of smooth muscle cells.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Chemokine CCL2/metabolism
- Disease Models, Animal
- Hypertension, Renovascular/etiology
- Hypertension, Renovascular/physiopathology
- Inflammation/etiology
- Inflammation/metabolism
- Inflammation/pathology
- Ligation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/pathology
- Myosin Heavy Chains/metabolism
- Plaque, Atherosclerotic/etiology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Renal Artery Obstruction/complications
- Vasculitis/etiology
- Vasculitis/metabolism
- Vasculitis/pathology
Collapse
Affiliation(s)
- G A Stouffer
- Division of Cardiology, University of North Carolina, Chapel Hill, NC 27599-7075, USA.
| | | | | |
Collapse
|
582
|
Meng L, Park J, Cai Q, Lanting L, Reddy MA, Natarajan R. Diabetic conditions promote binding of monocytes to vascular smooth muscle cells and their subsequent differentiation. Am J Physiol Heart Circ Physiol 2009; 298:H736-45. [PMID: 20008269 DOI: 10.1152/ajpheart.00935.2009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with significantly accelerated rates of atherosclerosis, key features of which include the presence of excessive macrophage-derived foam cells in the subendothelial space. We examined the hypothesis that enhanced monocyte-vascular smooth muscle cell (VSMC) interactions leading to subendothelial monocyte retention and differentiation to macrophages under diabetic conditions may be underlying mechanisms. Human aortic VSMCs (HVSMCs) treated with diabetic stimuli high glucose (HG) or S100B, a ligand of the receptor for advanced glycation end products, exhibited significantly increased binding of THP-1 monocytic cells. Diabetic stimuli increased the expression of the adhesive chemokine fractalkine (FKN) in HVSMCs. Pretreatment of HVSMCs with FKN or monocyte chemoattractant protein-1 (MCP-1) neutralizing antibodies significantly inhibited monocyte-VSMC binding, whereas monocytes treated with FKN showed enhanced binding to VSMC. Mouse aortic VSMCs (MVSMCs) derived from type 2 diabetic db/db mice exhibited significantly increased FKN levels and binding to mouse WEHI78/24 monocytic cells relative to nondiabetic control db/+ cells. The enhanced monocyte binding in db/db cells was abolished by both FKN and MCP-1 antibodies. Endothelium-denuded aortas from db/db mice and streptozotocin-induced diabetic mice also exhibited enhanced FKN expression and monocyte binding, relative to respective controls. Coculture with HVSMCs increased CD36 expression in THP-1 cells, and this was significantly augmented by treatment of HVSMCs with S100B or HG. CD36 mRNA and protein levels were also significantly increased in WEHI78/24 cells after coincubation with db/db MVSMCs relative to control MVSMCs. These results demonstrate that diabetic conditions may accelerate atherosclerosis by inducing key chemokines in the vasculature that promote VSMC-monocyte interactions, subendothelial monocyte retention, and differentiation.
Collapse
Affiliation(s)
- Li Meng
- Department of Diabetes, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | | | | | | | | | | |
Collapse
|
583
|
Roncal C, Buysschaert I, Gerdes N, Georgiadou M, Ovchinnikova O, Fischer C, Stassen JM, Moons L, Collen D, De Bock K, Hansson GK, Carmeliet P. Short-term delivery of anti-PlGF antibody delays progression of atherosclerotic plaques to vulnerable lesions. Cardiovasc Res 2009; 86:29-36. [PMID: 19952000 DOI: 10.1093/cvr/cvp380] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
AIMS Placental growth factor (PlGF), a homologue of vascular endothelial growth factor, is a pleiotropic cytokine with a pro-inflammatory activity. Previous gene-inactivation studies revealed that the loss of PlGF delays atherosclerotic lesion development and inhibits macrophage infiltration, but the activity of an anti-PlGF antibody (alphaPlGF mAb) has not been evaluated yet. METHODS AND RESULTS We characterized the potential of short-term delivery of alphaPlGF mAb in inhibiting lesion development in ApoE-deficient mice (apoE(-/-)) and in CD4:TGFbetaRII(DN) x apoE(-/-) mice, a more severe atherosclerosis model. Short-term treatment of alphaPlGF mAb reduces early atherosclerotic plaque size and inflammatory cell infiltration in the lesion. CONCLUSION These pharmacological alphaPlGF mAb results confirm previous genetic evidence that inhibition of PlGF slows down early atherosclerotic lesion development. Furthermore, the phenocopy of genetic and pharmacological loss-of-function strategies underscores that alphaPlGF acts by selectively neutralizing PlGF.
Collapse
Affiliation(s)
- Carmen Roncal
- Vesalius Research Center, VIB, KULeuven, Campus Gasthuisberg, Herestraat 49, Box 912, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
584
|
Ichikawa T, Li J, Dong X, Potts JD, Tang DQ, Li DS, Cui T. Ubiquitin carboxyl terminal hydrolase L1 negatively regulates TNFalpha-mediated vascular smooth muscle cell proliferation via suppressing ERK activation. Biochem Biophys Res Commun 2009; 391:852-6. [PMID: 19945429 DOI: 10.1016/j.bbrc.2009.11.151] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Accepted: 11/23/2009] [Indexed: 01/15/2023]
Abstract
Deubiquitinating enzymes (DUBs) appear to be critical regulators of a multitude of processes such as proliferation, apoptosis, differentiation, and inflammation. We have recently demonstrated that a DUB of ubiquitin carboxyl terminal hydrolase L1 (UCH-L1) inhibits vascular lesion formation via suppressing inflammatory responses in vasculature. However, the precise underlying mechanism remains to be defined. Herein, we report that a posttranscriptional up-regulation of UCH-L1 provides a negative feedback to tumor necrosis factor alpha (TNFalpha)-mediated activation of extracellular signal-regulated kinases (ERK) and proliferation in vascular smooth muscle cells (VSMCs). In rat adult VSMCs, adenoviral over-expression of UCH-L1 inhibited TNFalpha-induced activation of ERK and DNA synthesis. In contrast, over-expression of UCH-L1 did not affect platelet derived growth factor (PDGF)-induced VSMC proliferation and activation of growth stimulating cascades including ERK. TNFalpha hardly altered UCH-L1 mRNA expression and stability; however, up-regulated UCH-L1 protein expression via increasing UCH-L1 translation. These results uncover a novel mechanism by which UCH-L1 suppresses vascular inflammation.
Collapse
Affiliation(s)
- Tomonaga Ichikawa
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | | | | | | | | | | | | |
Collapse
|
585
|
Zhou Q, Liao JK. Rho kinase: an important mediator of atherosclerosis and vascular disease. Curr Pharm Des 2009; 15:3108-15. [PMID: 19754385 DOI: 10.2174/138161209789057986] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Atherosclerosis is a complex inflammatory process characterized by the cross-talk between excessive inflammation and lipid accumulation. In the past few years, compelling evidence suggests that statins can decrease vascular inflammation and attenuate the development of atherosclerosis through their so-called "pleiotropic effects". These cholesterol-independent effects are predominantly due to their ability to inhibit isoprenoid synthesis. In particular, inhibition of geranylgeranylpyrophosphate synthesis leads to inhibition of Rho and its downstream target, Rho-kinase (ROCK). Thus, one of the beneficial effects of statin therapy could be due to inhibitory effects on ROCK. ROCK is involved in mediating diverse cellular functions such as smooth muscle contraction, cell migration and proliferation. While increased ROCK activity is associated with endothelial dysfunction, cerebral ischemia, coronary vasospasms and metabolic syndrome, the inhibition of ROCK by statins or selective ROCK inhibitors leads to up-regulation of endothelial nitric oxide synthase (eNOS), decreased vascular inflammation, and reduced atherosclerotic plaque formation. This review will focus on the impact of ROCK in cardiovascular disease and its contributory role to vascular inflammation and the atherosclerosis.
Collapse
Affiliation(s)
- Qian Zhou
- Vascular Medicine Research Unit, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
586
|
The synaptic proteins neurexins and neuroligins are widely expressed in the vascular system and contribute to its functions. Proc Natl Acad Sci U S A 2009; 106:20782-7. [PMID: 19926856 DOI: 10.1073/pnas.0809510106] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Unlike other neuronal counterparts, primary synaptic proteins are not known to be involved in vascular physiology. Here, we demonstrate that neurexins and neuroligins, which constitute large and complex families of fundamental players in synaptic activity, are produced and processed by endothelial and vascular smooth muscle cells throughout the vasculature. Moreover, they are dynamically regulated during vessel remodeling and form endogenous complexes in large vessels as well as in the brain. We used the chicken chorioallantoic membrane as a system to pursue functional studies and demonstrate that a monoclonal recombinant antibody against beta-neurexin inhibits angiogenesis, whereas exogenous neuroligin has a role in promoting angiogenesis. Finally, as an insight into the mechanism of action of beta-neurexin, we show that the anti-beta-neurexin antibody influences vessel tone in isolated chicken arteries. Our finding strongly supports the idea that even the most complex and plastic events taking place in the nervous system (i.e., synaptic activity) share molecular cues with the vascular system.
Collapse
|
587
|
Schilling T, Eder C. Non-selective cation channel activity is required for lysophosphatidylcholine-induced monocyte migration. J Cell Physiol 2009; 221:325-34. [DOI: 10.1002/jcp.21857] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
588
|
Bui QT, Prempeh M, Wilensky RL. Atherosclerotic plaque development. Int J Biochem Cell Biol 2009; 41:2109-13. [DOI: 10.1016/j.biocel.2009.06.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 05/29/2009] [Accepted: 06/03/2009] [Indexed: 10/20/2022]
|
589
|
Orr AW, Hastings NE, Blackman BR, Wamhoff BR. Complex regulation and function of the inflammatory smooth muscle cell phenotype in atherosclerosis. J Vasc Res 2009; 47:168-80. [PMID: 19851078 DOI: 10.1159/000250095] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 08/26/2009] [Indexed: 12/22/2022] Open
Abstract
Vascular smooth muscle cell (SMC) phenotypic modulation plays a key role in atherosclerosis and is classically defined as a switch from a 'contractile' phenotype to a 'synthetic' phenotype, whereby genes that define the contractile SMC phenotype are suppressed and proliferation and/or migratory mechanisms are induced. There is also evidence that SMCs may take on a 'proinflammatory' phenotype, whereby SMCs secrete cytokines and express cell adhesion molecules, e.g. IL-8, IL-6, and VCAM-1, respectively, which may functionally regulate monocyte and macrophage adhesion and other processes during atherosclerosis. Factors that drive the inflammatory phenotype are not limited to cytokines but also include hemodynamic forces imposed on the blood vessel wall and intimate interaction of endothelial cells with SMCs, as well as changes in matrix composition in the vessel wall. However, it is critical to recognize that our understanding of the complex interaction of these multiple signal inputs has only recently begun to shed light on mechanisms that regulate the inflammatory SMC phenotype, primarily through models that attempt to recreate this environment ex vivo. The goal of this review is to summarize our current knowledge in this area and identify some of the key unresolved challenges and questions requiring further study.
Collapse
Affiliation(s)
- Anthony Wayne Orr
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, La., USA
| | | | | | | |
Collapse
|
590
|
Ahn SY, Choi YS, Koo HJ, Jeong JH, Park WH, Kim M, Piao Y, Pak YK. Mitochondrial dysfunction enhances the migration of vascular smooth muscles cells via suppression of Akt phosphorylation. Biochim Biophys Acta Gen Subj 2009; 1800:275-81. [PMID: 19781600 DOI: 10.1016/j.bbagen.2009.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 09/11/2009] [Accepted: 09/15/2009] [Indexed: 12/21/2022]
Abstract
BACKGROUND Atherosclerosis is one of the major complications of diabetes, which may result from insulin resistance via mitochondrial dysfunction. Although a strong association between insulin resistance and cardiovascular disease has been suggested, it is not clear yet whether stress-inducing factors damage mitochondria and insulin signaling pathway in cardiovascular tissues. METHODS We investigated whether stress-induced mitochondrial dysfunction might alter the insulin/Akt signaling pathway in A10 rat vascular smooth muscle cells (VSMC). RESULTS The treatment of oxidized low density lipoprotein (oxLDL) decreased ATP contents, mitochondrial respiration activity, mRNA expressions of OXPHOS subunits and IRS-1/2 and insulin-mediated phosphorylations of Akt and AMP-activated protein kinase (AMPK). Similarly, dideoxycytidine (ddC), the mtDNA replication inhibitor, or rotenone, OXPHOS complex I inhibitor, inhibited the insulin-mediated pAkt while increased pAMPK regardless of insulin. Reciprocally, an inhibitor of Akt, triciribine (TCN), decreased cellular ATP contents. Overexpression of Akt dominant positive reversed the oxLDL- or ddC-mediated ATP decrease but AMPK activator did not. Akt activation also normalized the aberrant VSMC migration induced by ddC. CONCLUSIONS Defective insulin signaling and mitochondrial function may collectively contribute to developing cardiovascular disease. GENERAL SIGNIFICANCE Akt may be a possible therapeutic target for treating insulin resistance-associated atherosclerosis.
Collapse
Affiliation(s)
- Sun Young Ahn
- Age-Related and Brain Diseases Research Center, Department of Nanopharmaceutical and Life Sciences, Department of Physiology, Kyung Hee University College of Medicine, Dongdaemoon-Gu Hoegi-Dong #1, Seoul 130-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
591
|
Boettger T, Beetz N, Kostin S, Schneider J, Krüger M, Hein L, Braun T. Acquisition of the contractile phenotype by murine arterial smooth muscle cells depends on the Mir143/145 gene cluster. J Clin Invest 2009; 119:2634-47. [PMID: 19690389 PMCID: PMC2735940 DOI: 10.1172/jci38864] [Citation(s) in RCA: 550] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 07/08/2009] [Indexed: 01/08/2023] Open
Abstract
VSMCs respond to changes in the local environment by adjusting their phenotype from contractile to synthetic, a phenomenon known as phenotypic modulation or switching. Failure of VSMCs to acquire and maintain the contractile phenotype plays a key role in a number of major human diseases, including arteriosclerosis. Although several regulatory circuits that control differentiation of SMCs have been identified, the decisive mechanisms that govern phenotypic modulation remain unknown. Here, we demonstrate that the mouse miR-143/145 cluster, expression of which is confined to SMCs during development, is required for VSMC acquisition of the contractile phenotype. VSMCs from miR-143/145-deficient mice were locked in the synthetic state, which incapacitated their contractile abilities and favored neointimal lesion development. Unbiased high-throughput, quantitative, mass spectrometry-based proteomics using reference mice labeled with stable isotopes allowed identification of miR-143/145 targets; these included angiotensin-converting enzyme (ACE), which might affect both the synthetic phenotype and contractile functions of VSMCs. Pharmacological inhibition of either ACE or the AT1 receptor partially reversed vascular dysfunction and normalized gene expression in miR-143/145-deficient mice. We conclude that manipulation of miR-143/145 expression may offer a new approach for influencing vascular repair and attenuating arteriosclerotic pathogenesis.
Collapse
Affiliation(s)
- Thomas Boettger
- Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany.
Albert-Ludwigs-Universität Freiburg, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany
| | - Nadine Beetz
- Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany.
Albert-Ludwigs-Universität Freiburg, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany
| | - Sawa Kostin
- Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany.
Albert-Ludwigs-Universität Freiburg, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany
| | - Johanna Schneider
- Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany.
Albert-Ludwigs-Universität Freiburg, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany
| | - Marcus Krüger
- Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany.
Albert-Ludwigs-Universität Freiburg, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany
| | - Lutz Hein
- Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany.
Albert-Ludwigs-Universität Freiburg, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany
| | - Thomas Braun
- Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany.
Albert-Ludwigs-Universität Freiburg, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany
| |
Collapse
|
592
|
Kim HJ, Yoo EK, Kim JY, Choi YK, Lee HJ, Kim JK, Jeoung NH, Lee KU, Park IS, Min BH, Park KG, Lee CH, Aronow BJ, Sata M, Lee IK. Protective role of clusterin/apolipoprotein J against neointimal hyperplasia via antiproliferative effect on vascular smooth muscle cells and cytoprotective effect on endothelial cells. Arterioscler Thromb Vasc Biol 2009; 29:1558-64. [PMID: 19696405 DOI: 10.1161/atvbaha.109.190058] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Clusterin is induced in vascular smooth muscle cells (VSMCs) during atherosclerosis and injury-induced neointimal hyperplasia. However, its functional roles in VSMCs and endothelial cells remain controversial and elusive. This study was undertaken to clarify the role of clusterin in neointimal hyperplasia and elucidate its mechanism of action. METHODS AND RESULTS Adenovirus-mediated overexpression of clusterin (Ad-Clu) repressed TNF-alpha-stimulated expression of MCP-1, fractalkine, ICAM-1, VCAM-1, and MMP-9, leading to inhibition of VSMC migration. Both Ad-Clu and secreted clusterin suppressed VSMC proliferation by inhibiting DNA synthesis, but not by inducing apoptosis. Ad-Clu upregulated p53 and p21(cip1/waf1) but downregulated cyclins D and E, leading to suppression of pRb phosphorylation and subsequent induction of G1 arrest in VSMCs. Clusterin deficiency augmented VSMC proliferation in vitro and accelerated neointimal hyperplasia in vivo, but concomitantly impaired reendothelialization in wire-injured murine femoral arteries. Moreover, Ad-Clu significantly reduced neointimal thickening in balloon-injured rat carotid arteries. Clusterin also diminished TNF-alpha-induced apoptosis of human umbilical vein endothelial cells and restored endothelial nitric oxide synthase expression suppressed by TNF-alpha. CONCLUSIONS These results suggest that upregulation of clusterin during vascular injury may be a protective response against, rather than a causative response to, the development of neointimal hyperplasia.
Collapse
Affiliation(s)
- Han-Jong Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 700-721, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
593
|
Riendeau V, Garenc C. Effect of 27-hydroxycholesterol on survival and death of human macrophages and vascular smooth muscle cells. Free Radic Res 2009; 43:1019-28. [PMID: 19672739 DOI: 10.1080/10715760903040610] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The objective was to compare the effect of a LXR synthetic ligand (T0901317) on cell viability and lysosomal membrane destabilization in human U937 macrophage and aortic smooth muscle cell (HASMC) incubated in the presence of cholesterol or 27-OH and to verify whether the Akt signalling pathway is involved. In U937 macrophages, cholesterol triggered cell survival while 27-OH triggered either survival (low concentration) or a lysosomal independent apoptosis (high concentration). Despite a strong effect of T0901317 on macrophage survival, its effect on cell viability is hampered in cells incubated in the presence of cholesterol or 27-OH. In these cells, cholesterol triggers the phosphorylation of Akt on the Thr308 residue. In HASMC, cholesterol induced apoptosis but no additionnal effect of T0901317 prevented apoptosis. All together, cell survival triggered by LXRs is impaired in the presence of cholesterol or high concentrations of 27-OH in human U937 macrophages and is not effective in HASMC.
Collapse
Affiliation(s)
- Valérie Riendeau
- Centre de recherche sur les maladies lipidiques, Centre hospitalier universitaire de Quebec/Centre de recherche du centre hospitalier de l'Universite Laval, Universite Laval, Quebec, QC, Canada
| | | |
Collapse
|
594
|
XXVIII Italian Society for the Study of Connective Tissues (SISC) Meeting, Pavia, Italy, 6–7 November 2008. Connect Tissue Res 2009. [DOI: 10.1080/03008200802683187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
595
|
Hsieh HL, Tung WH, Wu CY, Wang HH, Lin CC, Wang TS, Yang CM. Thrombin induces EGF receptor expression and cell proliferation via a PKC(delta)/c-Src-dependent pathway in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2009; 29:1594-601. [PMID: 19628787 DOI: 10.1161/atvbaha.109.185801] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Thrombin upregulates expression of several proteins in vascular smooth muscle cells (VSMCs) which may contribute to atherosclerosis. Here, we investigated the mechanisms underlying thrombin-induced EGF receptor (EGFR) expression and its effect on VSMCs. METHODS AND RESULTS Normal rat VSMCs were used. First, Western blotting and RT-PCR analyses showed that thrombin induces the expression of EGFR at transcription and translation levels in VSMCs. Second, pharmacological inhibitors, dominant negative mutants, and short hairpin RNA interference (shRNA) technology enabled us to demonstrate that thrombin-induced EGFR expression is mediated through PKC(delta)/c-Src-dependent transactivation of EGFR linking to PI3K/Akt and ERK1/2. We further investigated whether the transcription factors AP-1 and NF-kappaB are involved in this response by a promoter assay. Finally, data obtained by using EGFR shRNA technology and XTT assay demonstrated that thrombin-enhanced VSMC proliferation was mediated through upregulation of EGFR. CONCLUSIONS Our results demonstrate that thrombin-enhanced VSMC proliferation was mediated through upregulation of EGFR via a PKC(delta)/c-Src-dependent transactivation of EGFR, PI3K-Akt, and ERK, and AP-1/NF-kappaB pathway.
Collapse
Affiliation(s)
- Hsi-Lung Hsieh
- Department of Nursing, Division of Basic Medical Sciences, Chang Gung Institute of Technology, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
596
|
Substance P scavenger enhances antioxidant defenses and prevents prothrombotic effects on the rat lung after acute exposure to oil smoke. J Biomed Sci 2009; 16:58. [PMID: 19575822 PMCID: PMC2717930 DOI: 10.1186/1423-0127-16-58] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 07/06/2009] [Indexed: 11/29/2022] Open
Abstract
Background Airborne particulate matter, from cooking oil, smoking, engine exhaust and other sources, is associated with the development of atherosclerosis and myocardial infarction. In order to explore the cellular and molecular events following exposure of rats to lard oil smoke, we measured the generation of reactive oxygen species (ROS), substance P, cellular adhesion molecules, and thrombosis in relation to inhibitors of substance P, the NK-1 receptor, and antioxidants. Methods Rats were exposed to oil smoke for 120 min with or without 20 min pretreatment with lovastatin (substance P scavenger), L733060 (NK-1 receptor antagonist), vitamin E (antioxidant) or catechins (antioxidant). The levels of substance P and ROS were measured. Histological studies observed ROS damage in the form of HEL adducts. The prothrombotic effects of oil smoke exposure were measured by experimental induction of thrombosis in vivo. Results Oil smoke exposure significantly increased substance P levels, ROS levels, ROS damage (HEL adduct levels), and the size of experimentally induced thrombi. The pretreatments reduced all of these effects of oil smoke exposure; at many time points the reductions were statistically significant. Conclusion We established a connection between oil smoke exposure and thrombosis which involves substance P and its receptor, the NK-1 receptor, and ROS. This study helps establish a mechanistic explanation of how airborne particulate matter can increase the risk of cardiovascular illness.
Collapse
|
597
|
Lyle AN, Deshpande NN, Taniyama Y, Seidel-Rogol B, Pounkova L, Du P, Papaharalambus C, Lassègue B, Griendling KK. Poldip2, a novel regulator of Nox4 and cytoskeletal integrity in vascular smooth muscle cells. Circ Res 2009; 105:249-59. [PMID: 19574552 DOI: 10.1161/circresaha.109.193722] [Citation(s) in RCA: 348] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE NADPH oxidases (Noxes) regulate vascular physiology and contribute to the pathogenesis of vascular disease. In vascular smooth muscle cells (VSMCs), the interactions of individual Nox homologs with regulatory proteins are poorly defined. OBJECTIVE The objective of this study was to identify novel NADPH oxidase regulatory proteins. METHODS AND RESULTS Using a yeast 2-hybrid screen, we identified a novel p22phox binding partner, Poldip2, and demonstrated that it associates with p22phox, NADPH oxidase (Nox)1, and Nox4 and colocalizes with p22phox at sites of Nox4 localization. Poldip2 increases Nox4 enzymatic activity by 3-fold and positively regulates basal reactive oxygen species production in VSMCs (O2(.-): 86.3+/-15.6% increase; H2O2: 40.7+/-4.5% increase). Overexpression of Poldip2 activates Rho (180.2+/-24.8% increase), strengthens focal adhesions, and increases stress fiber formation. These phenotypic changes are blocked by dominant negative Rho. In contrast, depletion of either Poldip2 or Nox4 results in a loss of these structures, which is rescued by adding back active Rho. Cell migration, which requires dynamic cytoskeletal remodeling, is impaired by either excess (70.1+/-14.7% decrease) or insufficient Poldip2 (63.5+/-5.9% decrease). CONCLUSIONS These results suggest that Poldip2 associates with p22phox to activate Nox4, leading to regulation of focal adhesion turnover and VSMC migration, thus linking reactive oxygen species production and cytoskeletal remodeling. Poldip2 may be a novel therapeutic target for vascular pathologies with a significant VSMC migratory component, such as restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Alicia N Lyle
- Emory University, Division of Cardiology, 319 WMB, 1639 Pierce Dr, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
598
|
Kappert K, Meyborg H, Baumann B, Furundzija V, Kaufmann J, Graf K, Stibenz D, Fleck E, Stawowy P. Integrin cleavage facilitates cell surface-associated proteolysis required for vascular smooth muscle cell invasion. Int J Biochem Cell Biol 2009; 41:1511-7. [DOI: 10.1016/j.biocel.2009.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 12/22/2008] [Accepted: 01/05/2009] [Indexed: 11/16/2022]
|
599
|
Riou LM, Broisat A, Dimastromatteo J, Pons G, Fagret D, Ghezzi C. Pre-clinical and clinical evaluation of nuclear tracers for the molecular imaging of vulnerable atherosclerosis: an overview. Curr Med Chem 2009; 16:1499-511. [PMID: 19355903 DOI: 10.2174/092986709787909596] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of mortality worldwide. Despite major advances in the treatment of CVD, a high proportion of CVD victims die suddenly while being apparently healthy, the great majority of these accidents being due to the rupture or erosion of a vulnerable coronary atherosclerotic plaque. A non-invasive imaging methodology allowing the early detection of vulnerable atherosclerotic plaques in selected individuals prior to the occurrence of any symptom would therefore be of great public health benefit. Nuclear imaging could allow the identification of vulnerable patients by non-invasive in vivo scintigraphic imaging following administration of a radiolabeled tracer. The purpose of this review is to provide an overview of radiotracers that have been recently evaluated for the detection of vulnerable plaques together with the biological rationale that initiated their development. Radiotracers targeted at the inflammatory process seem particularly relevant and promising. Recently, macrophage targeting allowed the experimental in vivo detection of atherosclerosis using either SPECT or PET. A few tracers have also been evaluated clinically. Targeting of apoptosis and macrophage metabolism both allowed the imaging of vulnerable plaques in carotid vessels of patients. However, nuclear imaging of vulnerable plaques at the level of coronary arteries remains challenging, mostly because of their small size and their vicinity with unbound circulating tracer. The experimental and pilot clinical studies reviewed in the present paper represent a fundamental step prior to the evaluation of the efficacy of any selected tracer for the early, non-invasive detection of vulnerable patients.
Collapse
Affiliation(s)
- L M Riou
- INSERM, U877, Radiopharmaceutiques Biocliniques, Faculté de Médecine de Grenoble, F-38700, La tronche, France.
| | | | | | | | | | | |
Collapse
|
600
|
Hseu YC, Lin E, Chen JY, Liua YR, Huang CY, Lu FJ, Liao JW, Chen SC, Yang HL. Humic acid induces G1 phase arrest and apoptosis in cultured vascular smooth muscle cells. ENVIRONMENTAL TOXICOLOGY 2009; 24:243-258. [PMID: 18683188 DOI: 10.1002/tox.20426] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Humic acid (HA) in well water used by the inhabitants for drinking is one of the possible etiological factors for Blackfoot disease (BFD). In this study, the ability of HA to inhibit cell cycle progression and induce apoptosis in cultured smooth muscle cells (SMCs; A7r5) was investigated. Treatment of the SMCs at various HA concentrations (25-200 microg/mL) resulted in sequences of events marked by apoptosis, as shown by loss of cell viability, morphology change, and internucleosomal DNA fragmentation. HA-induced apoptotic cell death that is associated with loss of mitochondrial membrane potential (Delta Psi m), cytochrome c translocation, caspase-3, -8, and -9 activation, poly ADP-ribose polymerase (PARP) degradation, dysregulation of Bcl-2 and Bax, and upregulation of p53 and phospholyrated p53 (p-p53) in SMCs. Flow cytometry analysis demonstrated that HA blocked cell cycle progress in the G1 phase in SMCs. This blockade of cell cycle was associated with reduced amounts of cyclin D1, CDK4, cyclin E, CDK2, and hyperphosphorylated retinoblastoma protein (pRb) in a time-dependent manner. Apparent DNA strand breaks (DNA damage) were also detected in a dose-dependent manner using Single-cell gel electrophoresis assay (comet assay). Furthermore, HA induced dose-dependent elevation of reactive oxygen species (ROS) level in SMCs, and antioxidant vitamin C and Trolox effectively suppressed HA-induced DNA damage and dysregulation of Bcl-2/Bax. Our findings suggest that HA-induced DNA damage, cell cycle arrest, and apoptosis in SMCs may be an underlying mechanisms for the atherosclerosis and thrombosis observed in the BFD endemic region.
Collapse
Affiliation(s)
- You-Cheng Hseu
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|