1
|
Yuan Y, Zhou X, Wang Y, Wang Y, Teng X, Wang S. Cardiovascular Modulating Effects of Magnolol and Honokiol, Two Polyphenolic Compounds from Traditional Chinese Medicine-Magnolia Officinalis. Curr Drug Targets 2020; 21:559-572. [PMID: 31749425 DOI: 10.2174/1389450120666191024175727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 01/09/2023]
Abstract
Honokiol and its isomer magnolol are poly-phenolic compounds isolated from the Magnolia officinalis that exert cardiovascular modulating effects via a variety of mechanisms. They are used as blood-quickening and stasis-dispelling agents in Traditional Chinese Medicine and confirmed to have therapeutic potential in atherosclerosis, thrombosis, hypertension, and cardiac hypertrophy. This comprehensive review summarizes the current data regarding the cardioprotective mechanisms of those compounds and identifies areas for further research.
Collapse
Affiliation(s)
- Yuan Yuan
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaocui Zhou
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Yuanyuan Wang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Yan Wang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Xiangyan Teng
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Shuaiyu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
2
|
Chou CC, Wang CP, Chen JH, Lin HH. Anti-Atherosclerotic Effect of Hibiscus Leaf Polyphenols against Tumor Necrosis Factor-alpha-Induced Abnormal Vascular Smooth Muscle Cell Migration and Proliferation. Antioxidants (Basel) 2019; 8:antiox8120620. [PMID: 31817413 PMCID: PMC6943519 DOI: 10.3390/antiox8120620] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 01/21/2023] Open
Abstract
The proliferation and migration of vascular smooth muscle cells (VSMCs) are major events in the development of atherosclerosis following stimulation with proinflammatory cytokines, especially tumor necrosis factor-alpha (TNF-α). Plant-derived polyphenols have attracted considerable attention in the prevention of atherosclerosis. Hibiscus leaf has been showed to inhibit endothelial cell oxidative injury, low-density lipoprotein oxidation, and foam cell formation. In this study, we examined the anti-atherosclerotic effect of Hibiscus leaf polyphenols (HLPs) against abnormal VSMC migration and proliferation in vitro and in vivo. Firstly, VSMC A7r5 cells pretreated with TNF-α were demonstrated to trigger abnormal proliferation and affect matrix metalloproteinase (MMP) activities. Non-cytotoxic doses of HLPs abolished the TNF-α-induced MMP-9 expression and cell migration via inhibiting the protein kinase PKB (also known as Akt)/activator protein-1 (AP-1) pathway. On the other hand, HLP-mediated cell cycle G0/G1 arrest might be exerted by inducing the expressions of p53 and its downstream factors that, in turn, suppress cyclin E/cdk2 activity, preventing retinoblastoma (Rb) phosphorylation and the subsequent dissociation of Rb/E2F complex. HLPs also attenuated reactive oxygen species (ROS) production against TNF-α stimulation. In vivo, HLPs improved atherosclerotic lesions, and abnormal VSMC migration and proliferation. Our data present the first evidence of HLPs as an inhibitor of VSMC dysfunction, and provide a new mechanism for its anti-atherosclerotic activity.
Collapse
Affiliation(s)
- Cheng-Chung Chou
- Laboratory Medicine, Antai Medical Care Corporation Antai Tian-Sheng Memorial Hospital, Pingtung County 928, Taiwan;
| | - Chi-Ping Wang
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 0201, Taiwan;
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan
| | - Jing-Hsien Chen
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 0201, Taiwan;
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan
- Correspondence: (J.-H.C.); (H.-H.L.); Tel.: +886-424-730-022 (ext. 12195) (J.-H.C.); +886-424-730-022 (ext. 12410) (H.-H.L.)
| | - Hui-Hsuan Lin
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 0201, Taiwan;
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan
- Correspondence: (J.-H.C.); (H.-H.L.); Tel.: +886-424-730-022 (ext. 12195) (J.-H.C.); +886-424-730-022 (ext. 12410) (H.-H.L.)
| |
Collapse
|
3
|
Ho KJ, Owens CD. Diagnosis, classification, and treatment of femoropopliteal artery in-stent restenosis. J Vasc Surg 2017; 65:545-557. [PMID: 28126181 DOI: 10.1016/j.jvs.2016.09.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/13/2016] [Indexed: 01/19/2023]
Abstract
In-stent restenosis is a pervasive challenge to the durability of stenting for the treatment of lower extremity ischemia. There is considerable controversy about the criteria for diagnosis, indications for treatment, and preferred algorithm for addressing in-stent restenosis. This evidence summary seeks to review existing information on strategies for the treatment of this difficult problem.
Collapse
Affiliation(s)
- Karen J Ho
- Division of Vascular Surgery, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| | - Christopher D Owens
- Division of Vascular and Endovascular Surgery, University of California, San Francisco, Calif
| |
Collapse
|
4
|
Translational initiatives in thrombolytic therapy. Front Med 2017; 11:1-19. [DOI: 10.1007/s11684-017-0497-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 10/10/2016] [Indexed: 01/26/2023]
|
5
|
Swirski FK, Robbins CS, Nahrendorf M. Development and Function of Arterial and Cardiac Macrophages. Trends Immunol 2016; 37:32-40. [PMID: 26748179 DOI: 10.1016/j.it.2015.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/12/2015] [Accepted: 11/12/2015] [Indexed: 12/15/2022]
Abstract
Macrophages inhabit all major organs, and are capable of adapting their functions to meet the needs of their home tissues. The recent recognition that tissue macrophages derive from different sources, coupled with the notion that environmental cues and inflammatory stimuli can sculpt and agitate homeostasis, provides a frame of reference from which we can decipher the breadth and depth of macrophage activity. Here we discuss macrophages residing in the cardiovascular system, focusing particularly on their development and function in steady state and disease. Central to our discussion is the tension between macrophage ontogeny as a determinant of macrophage function, and the idea that tissues condition macrophage activities and supplant the influence of macrophage origins in favor of environmental demands.
Collapse
Affiliation(s)
- Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Clinton S Robbins
- Department of Immunology, Toronto General Research Institute, Peter Munk Cardiac Centre, University Health Network and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
6
|
Chan KC, Yu MH, Lin MC, Huang CN, Chung DJ, Lee YJ, Wu CH, Wang CJ. Pleiotropic effects of acarbose on atherosclerosis development in rabbits are mediated via upregulating AMPK signals. Sci Rep 2016; 6:38642. [PMID: 27924924 PMCID: PMC5141573 DOI: 10.1038/srep38642] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/11/2016] [Indexed: 01/10/2023] Open
Abstract
Acarbose, an α-glucosidase inhibitor, is reported to reduce the incidence of silent myocardial infarction and slow the progression of intima-media thickening in patients with glucose intolerance. Here we investigate other impacts of acarbose on atherosclerosis development and the underlying mechanisms of atherosclerosis initiation and progression in vivo and in vitro. Rabbits fed a high cholesterol diet (HCD) were treated with acarbose (2.5-5.0 mg kg-1). Immunohistochemistry was used to assess the expression of inducible nitric oxide synthase (iNOS), Ras, proliferating cell nuclear antigen (PCNA), IL-6, β-galactosidase, and p-AMPK in atherosclerotic lesions. Treatment with acarbose in HCD-fed rabbits was found to significantly reduce the severity of aortic atheroma and neointimal expression of α-actin, PCNA, IL-6, TNF-α, Ras, and β-galactosidase; to significantly increase expression of iNOS and p-AMPK, but not to affect serum levels of glucose, total cholesterol, and LDL. Western blot analysis showed acarbose dose-dependently decreased β-galactosidase and Ras expression and increased p-AMPK expression in TNF-α-treated A7r5 cells. In addition, acarbose restored p-AMPK and iNOS levels in AMPK inhibitor- and iNOS inhibitor-treated A7r5 cells, respectively. In conclusion, acarbose can pleiotropically inhibit rabbit atherosclerosis by reducing inflammation, senescence, and VSMCs proliferation/migration via upregulating AMPK signals.
Collapse
Affiliation(s)
- Kuei-Chuan Chan
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan.,School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan
| | - Meng-Hsun Yu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Ming-Cheng Lin
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan.,School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan
| | - Chien-Ning Huang
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan.,School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan
| | - Dai-Jung Chung
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Yi-Ju Lee
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan.,Department of Pathology, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan
| | - Cheng-Hsun Wu
- Department of Anatomy, China Medical University, Taichung, 404, Taiwan
| | - Chau-Jong Wang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung 402, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Road, Taichung, 402, Taiwan
| |
Collapse
|
7
|
Abstract
Diet, exercise, stress, and sleep are receiving attention as environmental modifiers of chronic inflammatory diseases, including atherosclerosis, the culprit condition of myocardial infarction and stroke. Accumulating data indicate that psychosocial stress and a high-fat, high-cholesterol diet aggravate cardiovascular disease, whereas regular physical activity and healthy sleeping habits help prevent it. Here, we raise the possibility that inflammation-associated leukocyte production plays a causal role in lifestyle effects on atherosclerosis progression. Specifically, we explore whether and how potent real-life disease modifiers influence hematopoiesis' molecular and cellular machinery. Lifestyle, we hypothesize, may rearrange hematopoietic topography, diverting production from the bone marrow to the periphery, thus propagating a quantitative and qualitative drift of the macrophage supply chain. These changes may involve progenitor-extrinsic and intrinsic communication nodes that connect organ systems along neuroimmune and immunometabolic axes, ultimately leading to an altered number and phenotype of lesional macrophages. We propose that, in conjunction with improved public health policy, future therapeutics could aim to modulate the quantitative and qualitative output, as well as the location, of the hematopoietic tree to decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston.
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston.
| |
Collapse
|
8
|
Loss of One Copy of Zfp148 Reduces Lesional Macrophage Proliferation and Atherosclerosis in Mice by Activating p53. Circ Res 2014; 115:781-9. [DOI: 10.1161/circresaha.115.304992] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Rationale:
Cell proliferation and cell cycle control mechanisms are thought to play central roles in the pathogenesis of atherosclerosis. The transcription factor Zinc finger protein 148 (Zfp148) was shown recently to maintain cell proliferation under oxidative conditions by suppressing p53, a checkpoint protein that arrests proliferation in response to various stressors. It is established that inactivation of p53 accelerates atherosclerosis, but whether increased p53 activation confers protection against the disease remains to be determined.
Objective:
We aimed to test the hypothesis that
Zfp148
deficiency reduces atherosclerosis by unleashing p53 activity.
Methods and Results:
Mice harboring a gene-trap mutation in the
Zfp148
locus (
Zfp148
gt/+
) were bred onto the apolipoprotein E (
Apoe
)
–/–
genetic background and fed a high-fat or chow diet. Loss of 1 copy of
Zfp148
markedly reduced atherosclerosis without affecting lipid metabolism. Bone marrow transplantation experiments revealed that the effector cell is of hematopoietic origin. Peritoneal macrophages and atherosclerotic lesions from
Zfp148
gt/+
Apoe
–/–
mice showed increased levels of phosphorylated p53 compared with controls, and atherosclerotic lesions contained fewer proliferating macrophages.
Zfp148
gt/+
Apoe
–/–
mice were further crossed with p53-null mice (
Trp53
–/–
[the gene encoding p53]). There was no difference in atherosclerosis between
Zfp148
gt/+
Apoe
–/–
mice and controls on a
Trp53
+/–
genetic background, and there was no difference in levels of phosphorylated p53 or cell proliferation.
Conclusions:
Zfp148
deficiency increases p53 activity and protects against atherosclerosis by causing proliferation arrest of lesional macrophages, suggesting that drugs targeting macrophage proliferation may be useful in the treatment of atherosclerosis.
Collapse
|
9
|
Kurakula K, Vos M, Otermin Rubio I, Marinković G, Buettner R, Heukamp LC, Stap J, de Waard V, van Tiel CM, de Vries CJ. The LIM-only protein FHL2 reduces vascular lesion formation involving inhibition of proliferation and migration of smooth muscle cells. PLoS One 2014; 9:e94931. [PMID: 24736599 PMCID: PMC3988136 DOI: 10.1371/journal.pone.0094931] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 03/20/2014] [Indexed: 01/01/2023] Open
Abstract
The LIM-only protein FHL2, also known as DRAL or SLIM3, has a function in fine-tuning multiple physiological processes. FHL2 is expressed in the vessel wall in smooth muscle cells (SMCs) and endothelial cells and conflicting data have been reported on the regulatory function of FHL2 in SMC phenotype transition. At present the function of FHL2 in SMCs in vascular injury is unknown. Therefore, we studied the role of FHL2 in SMC-rich lesion formation. In response to carotid artery ligation FHL2-deficient (FHL2-KO) mice showed accelerated lesion formation with enhanced Ki67 expression compared with wild-type (WT)-mice. Consistent with these findings, cultured SMCs from FHL2-KO mice showed increased proliferation through enhanced phosphorylation of extracellular-regulated kinase-1/2 (ERK1/2) and induction of CyclinD1 expression. Overexpression of FHL2 in SMCs inhibited CyclinD1 expression and CyclinD1-knockdown blocked the enhanced proliferation of FHL2-KO SMCs. We also observed increased CyclinD1 promoter activity in FHL2-KO SMCs, which was reduced upon ERK1/2 inhibition. Furthermore, FHL2-KO SMCs showed enhanced migration compared with WT SMCs. In conclusion, FHL2 deficiency in mice results in exacerbated SMC-rich lesion formation involving increased proliferation and migration of SMCs via enhanced activation of the ERK1/2-CyclinD1 signaling pathway.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mariska Vos
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Iker Otermin Rubio
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Goran Marinković
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Jan Stap
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudia M. van Tiel
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlie J.M. de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
10
|
White RD, Holdaway BB, Moody JD, Chang Y. Chronic Caffeine Administration Attenuates Vascular Injury-Induced Neointimal Hyperplasia in Rats. JOURNAL OF CAFFEINE RESEARCH 2013; 3:163-168. [PMID: 24761282 DOI: 10.1089/jcr.2013.0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Inflammation is considered to be a major initiator to angioplasty-induced vascular restenosis. Proinflammatory cytokines stimulate vascular smooth muscle cell (VSMC) migration and proliferation leading to neointimal hyperplasia. It has been reported that chronic caffeine use suppresses the production of proinflammatory cytokine TNF-α (tumor necrosis factor Alpha) and alters adenosine receptor expression in human neutrophils, indicating that caffeine may attenuate vascular injury-induced inflammation and subsequent neointimal hyperplasia. Our current study was designed to test the hypothesis that chronic caffeine treatment decreases vascular injury-induced neointimal hyperplasia by suppressing VSMC migration and proliferation. Methods and Results: The experiments were carried out using both in vivo (rat carotid artery injury model) and in vitro (VSMCs isolated from rat aorta) models. Male Sprague-Dawley rats that received chronic caffeine treatment (10 and 20 mg/kg per day, through oral gavage) showed a significant decrease in neointimal hyperplasia when compared to rats that received vehicle. To understand the underlying mechanisms, we tested if caffeine inhibits fetal bovine serum (FBS)-induced VSMC migration and proliferation. We found that caffeine substantially suppressed FBS-induced VSMC migration and proliferation. The attenuation of FBS-stimulated cell migration is dose dependent. Conclusion: Together, our results suggest that chronic treatment with high concentrations of caffeine attenuates vascular injury-induced neointimal hyperplasia by suppressing smooth muscle cell migration and proliferation in rats.
Collapse
Affiliation(s)
- Ryan D White
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri. ; University of Missouri School of Medicine , Columbia, Missouri
| | - Brett B Holdaway
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri
| | - Joshua D Moody
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri
| | - Yingzi Chang
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences , Kirksville, Missouri
| |
Collapse
|
11
|
Hutter R, Huang L, Speidl WS, Giannarelli C, Trubin P, Bauriedel G, Klotman ME, Fuster V, Badimon JJ, Klotman PE. Novel small leucine-rich repeat protein podocan is a negative regulator of migration and proliferation of smooth muscle cells, modulates neointima formation, and is expressed in human atheroma. Circulation 2013; 128:2351-63. [PMID: 24043300 DOI: 10.1161/circulationaha.113.004634] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Smooth muscle cell (SMC) migration and proliferation critically influence the clinical course of vascular disease. We tested the effect of the novel small leucine-rich repeat protein podocan on SMC migration and proliferation using a podocan-deficient mouse in combination with a model of arterial injury and aortic explant SMC culture. In addition, we examined the effect of overexpression of the human form of podocan on human SMCs and tested for podocan expression in human atherosclerosis. In all these conditions, we concomitantly evaluated the Wnt-TCF (T-cell factor) pathway. METHODS AND RESULTS Podocan was strongly and selectively expressed in arteries of wild-type mice after injury. Podocan-deficient mice showed increased arterial lesion formation compared with wild-type littermates in response to injury (P<0.05). Also, SMC proliferation was increased in arteries of podocan-deficient mice compared with wild-type (P<0.05). In vitro, migration and proliferation were increased in podocan-deficient SMCs and were normalized by transfection with the wild-type podocan gene (P<0.05). In addition, upregulation of the Wnt-TCF pathway was found in SMCs of podocan-deficient mice both in vitro and in vivo. On the other hand, podocan overexpression in human SMCs significantly reduced SMC migration and proliferation, inhibiting the Wnt-TCF pathway. Podocan and a Wnt-TCF pathway marker were differently expressed in human coronary restenotic versus primary lesions. CONCLUSIONS Podocan appears to be a potent negative regulator of the migration and proliferation of both murine and human SMCs. The lack of podocan results in excessive arterial repair and prolonged SMC proliferation, which likely is mediated by the Wnt-TCF pathway.
Collapse
Affiliation(s)
- Randolph Hutter
- Departments of Medicine and Cardiology, Mount Sinai School of Medicine, New York, NY (R.H., L.H., W.S.S., C.G., P.T., V.F., J.J.B.); Department of Cardiology, Elisabeth Klinikum, Schmalkalden, Germany (G.B.); Department of Medicine, Duke University, Durham, NC (M.E.K.); and Department of Medicine, Baylor College of Medicine, Houston, TX (P.E.K.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
A novel model of intimal hyperplasia with graded hypoosmotic damage. Cardiovasc Pathol 2012; 21:490-8. [DOI: 10.1016/j.carpath.2012.02.007] [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] [Received: 12/02/2011] [Revised: 02/19/2012] [Accepted: 02/20/2012] [Indexed: 11/24/2022] Open
|
13
|
Abstract
Percutaneous transluminal coronary angioplasty is a widely used technique for recanalizing arteries that are occluded by atherosclerotic plaque, but its usefulness is limited by the occurrence ofrestenosis in a high proportion of patients. The development of new therapies for this currently intractable problem will be facilitated by the use of animal models of restenosis that are predictive of drug efficacy in humans. Two approaches for improving predictivity can be identified. In the first of these, the goal is to maximize the anatomical and procedural resemblance of the model to humans. The second approach seeks to maximize the pathophysiological and molecular biological resemblance of the model to humans. Tangible progress is being made toward the first goal, but lack of understanding of the basic biology of human restenosis is hampering progress toward the second.
Collapse
|
14
|
Beyea MM, Reaume S, Sawyez CG, Edwards JY, O'Neil C, Hegele RA, Pickering JG, Huff MW. The oxysterol 24(s),25-epoxycholesterol attenuates human smooth muscle-derived foam cell formation via reduced low-density lipoprotein uptake and enhanced cholesterol efflux. J Am Heart Assoc 2012; 1:e000810. [PMID: 23130136 PMCID: PMC3487330 DOI: 10.1161/jaha.112.000810] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 05/08/2012] [Indexed: 01/30/2023]
Abstract
Background Foam cell formation by intimal smooth muscle cells (SMCs) inhibits the elaboration of extracellular matrix, which is detrimental to plaque stabilization. In the present study, we examined the lipoproteins and receptors involved in human SMC foam cell formation and investigated the ability of 24(S),25-epoxycholesterol [24(S),25-EC], an oxysterol agonist of the liver X receptor, to attenuate SMC foam cell formation. Methods and Results Incubation of human internal thoracic SMCs with atherogenic lipoproteins demonstrated that low-density lipoprotein (LDL), but not oxidized or acetylated LDL, was the primary lipoprotein taken up, resulting in marked cholesteryl ester deposition (6-fold vs 1.8-fold; P<0.05; n=4). Exposure of SMCs to exogenous or endogenously synthesized 24(S),25-EC attenuated LDL uptake (−90% and −47% respectively; P<0.05; n=3) through decreased sterol regulatory element–binding protein-2 expression (−30% and −17%, respectively; P<0.001; n=3), decreased LDL receptor expression (−75% and −40%, respectively; P<0.05; n=3) and increased liver X receptor–mediated myosin regulatory light chain interacting protein expression (7- and 3-fold, respectively; P<0.05; n=4). Furthermore, exogenous 24(S),25-EC increased adenosine triphosphate–binding cassettes A1– and G1–mediated cholesterol efflux to apolipoprotein AI (1.9-fold; P<0.001; n=5) and high-density lipoprotein3 (1.3-fold; P<0.05; n=5). 24(S),25-EC, unlike a nonsteroidal liver X receptor agonist, T0901317, did not stimulate sterol regulatory element–binding protein-1c–mediated fatty acid synthesis or triglyceride accumulation. 24(S),25-EC preserved the assembly of fibronectin and type I collagen by SMCs. Conclusions The oxysterol 24(S),25-EC prevented foam cell formation in human SMCs by attenuation of LDL receptor–mediated LDL uptake and stimulation of cholesterol efflux, restoring the elaboration of extracellular matrix. In contrast to T0901317, 24(S),25-EC prevented the development of a triglyceride-rich foam cell phenotype. (J Am Heart Assoc. 2012;1:e000810 doi: 10.1161/JAHA.112.000810.)
Collapse
Affiliation(s)
- Michael M Beyea
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada (M.M.B., C.G.S., J.Y.E., C.O., R.A.H., J.G.P., M.W.H) ; Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada (M.M.B., S.R., R.A.H., J.G.P., M.W.H.)
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Interleukin (IL)-17 (also known as IL-17A) is produced by activated T cells. It is a marker cytokine of the T(H₁₇) lineage. IL-17 production is induced in infections, autoimmune diseases and other inflammatory events. IL-17 is involved in host defense, but also inflammatory tissue destruction. Vascular disease, mostly in the chronic form of atherosclerosis, is a leading cause of death. While normal vessels harbor only few leukocytes, large numbers of both innate and adaptive immune cells accumulate during vascular inflammation, both in chronic forms such as atherosclerosis and in acute vasculitis. IL-17 has a role in chronic vascular inflammation of atherosclerosis and possibly hypertensive vascular changes. In acute inflammation, IL-17 is elevated and may be causally involved in the autoimmune vasculitides including vasculitis in systemic lupus erythematodes. Blood vessels are important targets in alloimmune graft rejection and a number of studies provide data on a role of IL-17 in this context. This brief review summarizes the currently available evidence for and putative mechanisms of action of IL-17 in mouse models of and human vascular disease.
Collapse
Affiliation(s)
- Sibylle von Vietinghoff
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | |
Collapse
|
16
|
Yi I, Lee JJ, Park JS, Zhang WY, Kim IS, Kim Y, Shin CY, Kim HS, Myung CS. Enhanced effect of losartan and rosuvastatin on neointima hyperplasia. Arch Pharm Res 2010; 33:593-600. [PMID: 20422369 DOI: 10.1007/s12272-010-0414-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/11/2010] [Accepted: 01/24/2010] [Indexed: 10/19/2022]
|
17
|
Zettler ME, Merchant MA, Pierce GN. Augmented cell cycle protein expression and kinase activity in atherosclerotic rabbit vessels. Exp Clin Cardiol 2010; 15:e139-e144. [PMID: 21264072 PMCID: PMC3016074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Cell proliferation within a primary atherosclerotic plaque is controversial. Identifying changes in cell cycle protein expression and the activities of their related kinases would provide valuable evidence of mitotic activity in the atherosclerotic lesion. Oxidized low-density lipoprotein has been shown to induce a significant increase in the total number of rabbit vascular smooth muscle cells in culture. In the present study, whole aortic cell extracts were harvested from rabbits fed a cholesterol-supplemented diet for eight weeks to induce modest plaque development, or 16 weeks to induce later, more severe plaque progression. Expression levels of cyclin A, cyclin-dependent kinase 4 (Cdk 4) and proliferating cell nuclear antigen were measured, as well as the activities of Cdk 4, Cdk 2 and Cdk 1. At both time points, the expression levels of cyclin A, Cdk 4 and proliferating cell nuclear antigen were significantly elevated. The activity of all three Cdks was also increased. There were no significant differences between moderate and more severe atherosclerosis. Surprisingly, tissues that neighboured the plaques, but did not show visible plaque formation on the vessel surface, also had significantly elevated cyclin A expression levels, but not as high as in the plaque areas. In conclusion, the primary atherosclerotic plaque exhibited elevated mitotic activity as shown by increased expression levels and activities of several cell cycle proteins. Expression levels were similar during moderate and severe atherosclerosis, and were even detected in nonatherosclerotic vascular tissue bordering the plaque.
Collapse
Affiliation(s)
| | | | - Grant N Pierce
- Correspondence: Dr Grant N Pierce, St Boniface General Hospital Research Centre, 351 Tache Avenue, Winnipeg, Manitoba R2H 2A6. Telephone 204-235-3206, fax 204-235-0793, e-mail
| |
Collapse
|
18
|
Manolakou P, Angelopoulou R, Bakoyiannis C, Psathas E, Bastounis E, Kavantzas N, Patsouris E. Cellular proliferation in complicated versus uncomplicated atherosclerotic lesions: Total cell population, foam cells and newly formed microvessels. Tissue Cell 2009; 41:408-13. [DOI: 10.1016/j.tice.2009.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 05/20/2009] [Accepted: 05/25/2009] [Indexed: 11/29/2022]
|
19
|
Fuster JJ, Fernández P, González-Navarro H, Silvestre C, Nabah YNA, Andrés V. Control of cell proliferation in atherosclerosis: insights from animal models and human studies. Cardiovasc Res 2009; 86:254-64. [PMID: 19900964 DOI: 10.1093/cvr/cvp363] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Excessive hyperplastic cell growth within occlusive vascular lesions has been recognized as a key component of the inflammatory response associated with atherosclerosis, restenosis post-angioplasty, and graft atherosclerosis after coronary artery bypass. Understanding the molecular mechanisms that regulate arterial cell proliferation is therefore essential for the development of new tools for the treatment of these diseases. Mammalian cell proliferation is controlled by a large number of proteins that modulate the mitotic cell cycle, including cyclin-dependent kinases, cyclins, and tumour suppressors. The purpose of this review is to summarize current knowledge about the role of these cell cycle regulators in the development of native and graft atherosclerosis that has arisen from animal studies, histological examination of specimens from human patients, and genetic studies.
Collapse
Affiliation(s)
- José J Fuster
- Laboratory of Vascular Biology, Department of Molecular and Cellular Pathology and Therapy, Instituto de Biomedicina de Valencia-CSIC, C/Jaime Roig 11, 46010 Valencia, Spain
| | | | | | | | | | | |
Collapse
|
20
|
Characterization of primary and restenotic atherosclerotic plaque from the superficial femoral artery: Potential role of Smad3 in regulation of SMC proliferation. J Vasc Surg 2009; 49:1289-95. [PMID: 19394554 DOI: 10.1016/j.jvs.2008.11.096] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 08/25/2008] [Accepted: 11/25/2008] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To characterize and compare primary and restenotic lesions of the superficial femoral artery and analyze the contribution of TGF-beta/Smad3 signaling to the pathophysiology of peripheral artery occlusive disease. METHODS AND RESULTS Immunohistochemical studies were performed on specimens retrieved from the superficial femoral artery of patients undergoing either atherectomy for primary atherosclerotic or recurrent disease after stenting and/or prior angioplasty. Immunohistochemical analysis revealed a significantly higher smooth muscle cell (SMC) content (alpha-actin+) and expression of Smad3 in restenotic lesions while primary lesions contained significantly more leukocytes (CD45+) and macrophages (CD68+). Further studies demonstrated colocalization of Smad3 with alpha-actin and PCNA, suggesting a role for Smad3 in the proliferation observed in restenotic lesions. To confirm a role for Smad3 in SMC proliferation, we both upregulated Smad3 via adenoviral mediated gene transfer (AdSmad3) and inhibited Smad3 through transfection with siRNA in human aortic SMCs, then assessed cell proliferation with tritiated thymidine. Overexpression of Smad3 enhanced whereas inhibition of Smad3 decreased cell proliferation. CONCLUSION Differences in cellular composition and cell proliferation in conjunction with the finding that Smad3 is expressed exclusively in restenotic disease suggest that TGF-beta, through Smad3 signaling, may play an essential role in SMC proliferation and the pathophysiology of restenosis in humans.
Collapse
|
21
|
Abstract
The long term outcome of stent implantation is affected by a process called in stent restenosis (ISR). Multiple contributory factors have been identified, but clear understanding of the overall underlying mechanism remains an enigma. ISR progresses through several different phases and involves numerous cellular and molecular constituents. Platelets and macrophages play a central role via vascular smooth muscle cell migration and proliferation in the intima to produce neointimal hyperplasia, which is pathognomic of ISR. Increased extracellular matrix formation appears to form the bulk of the neointimal hyperplasia tissue. Emerging evidence of the role of inflammatory cytokines and suppressors of cytokine signalling make this an exciting and novel field of antirestenosis research. Activation of Akt pathway triggered by mechanical stretch may also be a contributory factor to ISR formation. Prevention of ISR appears to be a multipronged attack as no therapeutic "magic bullet" exists to block all the processes in one go.
Collapse
Affiliation(s)
- A K Mitra
- Departments of Biomedical Sciences, Medicine, and Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | |
Collapse
|
22
|
Liu SC, Wang SS, Wu MZ, Wu DC, Yu FJ, Chen WJ, Chiang FT, Yu MF. Activation of telomerase and expression of human telomerase reverse transcriptase in coronary atherosclerosis. Cardiovasc Pathol 2005; 14:232-40. [PMID: 16168895 DOI: 10.1016/j.carpath.2005.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 05/07/2005] [Accepted: 05/12/2005] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Considerable research on telomerase on human neoplastic and normal long-lived proliferative tissues has emerged. We explored the expression of telomerase in atherosclerotic human epicardial coronary arteries. METHODS Forty discrete human coronary arterial segments obtained from 19 heart transplant recipients were classified into nonatherosclerotic and atherosclerotic groups based on coronary angiography and histological examination. PCR-ELISA-based telomeric repeat amplification protocol (TRAP), and immunohistochemical analyses were conducted to determine the functional activity and cell-specific expression of telomerase. RESULTS Seventy percent of atherosclerotic coronary arteries exhibited positive telomerase activity, and the reactivation incidence reached fourfold higher than that of controls (P=.007). The telomerase catalytic protein, human telomerase reverse transcriptase (hTERT), was expressed in 88% of atherosclerotic tissues, a fivefold higher frequency compared with that of the controls. There was also a correlation of hTERT expression with the level of telomerase bioactivity (P=.017) and with the severity of atherosclerotic grade (P<.001). In comparison with the immunostaining of mitotic antigen, Ki-67, we found an association of hTERT expression with actively cycling cells in early lesions but with quiescent cells in late advanced atherosclerotic stages. CONCLUSIONS The up-regulation of telomerase and its catalytic hTERT protein during stages of atherosclerotic evolution may implicate a role of telomerase in vascular remodeling underlying atherogenesis.
Collapse
Affiliation(s)
- Shih-Chi Liu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The aim of this study was to reconstruct dynamic biological steps of human atherosclerosis at different ages of life and, in particular, to clarify the role of the smooth muscle cells (SMCs) by means of evaluation of several markers implicated in proliferative diseases (c-fos, proliferating cell nuclear antigen: PCNA, apoptosis, chromosome 7). We examined the biological features of 67 atherosclerotic arterial lesions obtained from fetuses, infants, young people and adults. From each case serial sections were stained for histological examination, PCNA, c-fos and apoptosis detection by immunohistochemical methods and for chromosome 7 number evaluation by fluorescence in situ hybridization. In coronary specimens of fetuses we observed SMCs with c-fos positivity. In infant lesions the predominant result was positivity for PCNA. Similar results were obtained from the plaques from young subjects with a greater presence of PCNA-positive cells. In adult subjects numerous apoptotic cells were present in the stable plaques, whereas in the unstable plaques we frequently detected joint positivity for both PCNA and c-fos gene and supernumerary chromosomes 7. During the evolution of the atherosclerotic process we observed a biological modulation of SMC proliferation, which begins after activation of the c-fos gene, increases during progression of the lesion and declines in stable plaques, when apoptosis increases. In unstable plaques, the same early steps observed in fetus and infant arteries occur. The observation in some cases of chromosome 7 alterations, markers of tumorigenesis, suggests the possible transformation of an advanced atherosclerotic plaque into a neoplastic-like process.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Apoptosis/physiology
- Child, Preschool
- Chromosome Aberrations
- Chromosomes, Human, Pair 7/genetics
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/pathology
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Female
- Fetus
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Infant
- Male
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proliferating Cell Nuclear Antigen/metabolism
- Proto-Oncogene Proteins c-fos/metabolism
Collapse
|
24
|
Edo MD, Roldán M, Andrés V. Cyclin-dependent protein kinases as therapeutic targets in cardiovascular disease. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.13.5.579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
25
|
Abstract
Worldwide, more people die of the complications of atherosclerosis than of any other cause. It is not surprising, therefore, that enormous resources have been devoted to studying the pathogenesis of this condition. This article attempts to summarize present knowledge on the events that take place within the arterial wall during atherogenesis. Classical risk factors are not dealt with as they are the subjects of other parts of this book. First, we deal with the role of endothelial dysfunction and infection in initiating the atherosclerotic lesion. Then we describe the development of the lesion itself, with particular emphasis on the cell types involved and the interactions between them. The next section of the chapter deals with the events leading to thrombotic occlusion of the atherosclerotic vessel, the cause of heart attack and stroke. Finally, we describe the advantages--and limitations--of current animal models as they contribute to our understanding of atherosclerosis and its complications.
Collapse
Affiliation(s)
- P Cullen
- Institute of Arteriosclerosis Research, Münster, Germany.
| | | | | |
Collapse
|
26
|
Abstract
The 2 major general concepts about the cell biology of atherogenesis, growth of smooth muscle cells, and lipid accumulation in macrophages, ie, foam cell formation, have not been able to satisfactorily explain the genesis of acute coronary syndromes. Rather, the basic pathology behind the acute atherothrombotic events relates to erosion and rupture of unstable coronary plaques. At the cellular level, we now understand that a switch from cellular growth to cellular death, notably apoptosis, could be involved in turning at least some types of atherosclerotic plaques unstable. Because intimal cells require a proper matrix environment for normal function and survival, the vulnerability of an atherosclerotic plaque may critically depend on the integrity of the pericellular matrix of the plaque cells. In vitro studies have revealed that plaque-infiltrating inflammatory cells, such as macrophages, T-lymphocytes, and mast cells, by secreting a variety of proteases capable of degrading pericellular matrix components, induce death of endothelial cells and smooth muscle cells, and so provide a mechanistic explanation for inflammation-dependent plaque erosion and rupture. Thus, a novel link between inflammation and acute coronary syndromes is emerging. For a more explicit understanding of the role of proteases released by inflammatory cells in the conversion of a clinically silent plaque into a dangerous and potentially killing plaque, animal models of plaque erosion and rupture need to be established.
Collapse
|
27
|
Chang CJ, Ko PJ, Hsu LA, Ko YS, Ko YL, Chen CF, Huang CC, Hsu TS, Lee YS, Pang JHS. Highly increased cell proliferation activity in the restenotic hemodialysis vascular access after percutaneous transluminal angioplasty: implication in prevention of restenosis. Am J Kidney Dis 2004; 43:74-84. [PMID: 14712430 DOI: 10.1053/j.ajkd.2003.09.015] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND The effect of percutaneous transluminal angioplasty (PTA) in the treatment of hemodialysis vascular access stenosis is attenuated by a high restenosis rate, which results mainly from neointimal hyperplasia. Cellular proliferation is one of the most important biological mechanisms involved in neointimal hyperplasia and may be a potential target of intervention to prevent restenosis. METHODS We investigated the activity of cellular proliferation of restenotic lesions by means of immunohistochemistry, using an antibody to the proliferating cell nuclear antigen. Specimens from 10 primary stenotic and 20 restenotic lesions of 30 Brescia-Cimino fistulae were obtained during revision. RESULTS The proliferation index of the restenotic group was strikingly significantly greater than that of the primary stenotic group (intima, P < 0.001; media, P = 0.001). Proliferation indices of patients with diabetes in the restenotic group were significantly higher than those of patients without diabetes (intima, P = 0.028; media, P = 0.002). In the restenotic group, proliferation indices correlated negatively with the interval from PTA to restenosis (intima, r = -0.741; P < 0.001; media, r = -0.589; P = 0.006) and positively with the number of PTAs per lesion (intima, r = 0.754; P < 0.001; media, r = 0.506; P = 0.004). CONCLUSION We show markedly high cellular proliferation activity in early restenotic lesions of arteriovenous fistulae. These findings indicate that adjunctive antiproliferative therapy is mandatory in preventing restenosis after PTA, especially in patients with diabetes.
Collapse
Affiliation(s)
- Chi-Jen Chang
- First Cardiovascular Division, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Schober A, Knarren S, Lietz M, Lin EA, Weber C. Crucial Role of Stromal Cell–Derived Factor-1α in Neointima Formation After Vascular Injury in Apolipoprotein E–Deficient Mice. Circulation 2003; 108:2491-7. [PMID: 14581398 DOI: 10.1161/01.cir.0000099508.76665.9a] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Recent evidence indicates that stromal cell–derived factor-1α (SDF-1α) is expressed in human atherosclerotic plaques, whereas high plasma levels are clinically associated with stable coronary artery disease. Herein, we investigate the involvement of SDF-1α in neointimal formation after vascular injury.
Methods and Results—
SDF-1α was detected by immunohistochemistry in carotid arteries of apolipoprotein E–deficient (apoE
−/−
) mice at various stages of neointima formation after wire-induced injury. Double immunofluorescence revealed that SDF-1α staining was mostly confined to smooth muscle cells (SMCs). Furthermore, SDF-1α plasma levels peaked 1 day after vascular injury. Treatment of apoE
−/−
mice after carotid injury with a neutralizing SDF-1α monoclonal antibody for 3 weeks reduced neointimal lesion area by 44% (n=5,
P
<0.05) compared with isotype control. In SDF-1α antibody–treated apoE
−/−
mice, neointimal SMC content was decreased (21.7±2% versus 39.4±4%, n=5,
P
=0.005), whereas the relative content of neointimal macrophages remained unchanged. As shown by flow cytometry, carotid injury resulted in a marked expansion of circulating Sca-1
+
lineage
−
progenitor cells (PBPCs) in the peripheral blood of apoE
−/−
mice after 1 day, which was mediated by SDF-1α. Systemic injection of isolated PBPCs after vascular injury demonstrated their recruitment to neointimal lesions, where they can adopt an SMC-like phenotype.
Conclusions—
SDF-1α plays an instrumental role in neointimal formation after vascular injury in apoE
−/−
mice by regulating neointimal SMC content. This contribution appears to be attributable to SDF-1α–dependent recruitment of circulating SMC progenitor cells.
Collapse
Affiliation(s)
- Andreas Schober
- Department of Molecular Cardiovascular Research, University Hospital, Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | | | | | | | | |
Collapse
|
29
|
Mnjoyan ZH, Fujise K. Profound negative regulatory effects by resveratrol on vascular smooth muscle cells: a role of p53–p21WAF1/CIP1 pathway. Biochem Biophys Res Commun 2003; 311:546-52. [PMID: 14592451 DOI: 10.1016/j.bbrc.2003.10.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We investigated the role of resveratrol, a polyphenol rich in red wine, in cell cycle progression and apoptosis of vascular smooth muscle cells (VSMCs). Resveratrol inhibited the growth of human aortic VSMCs at concentrations as low as 1 microM. This was due to the profound dose-dependent inhibition of DNA synthesis by resveratrol. DNA synthesis was more effectively inhibited when cells were pretreated with resveratrol. Resveratrol caused a dose-dependent increase in intracellular p53 and p21(WAF1/CIP1) levels. At lower concentrations (6.25-12.5 microM), resveratrol effectively blocked cell cycle progression of serum-stimulated VSMCs without inducing apoptosis, while the higher concentration of resveratrol (25 microM) selectively induced apoptosis in the same VSMCs. Intriguingly, however, the same high concentration of resveratrol could not induce apoptosis in quiescent VSMCs. These differential biological effects of resveratrol on quiescent and proliferating VSMCs suggest that resveratrol may be capable of selectively eliminating abnormally proliferating VSMCs of the arterial walls in vivo.
Collapse
MESH Headings
- Aorta/anatomy & histology
- Aorta/drug effects
- Aorta/growth & development
- Aorta/metabolism
- Apoptosis/drug effects
- Cell Division/drug effects
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p21
- Cyclins/metabolism
- DNA Replication/drug effects
- Dose-Response Relationship, Drug
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Resveratrol
- Stilbenes/metabolism
- Stilbenes/pharmacology
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Zakar H Mnjoyan
- Research Center for Cardiovascular Diseases, Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | |
Collapse
|
30
|
Chen JH, Wu CC, Hsiao G, Yen MH. Magnolol induces apoptosis in vascular smooth muscle. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2003; 368:127-33. [PMID: 12898128 DOI: 10.1007/s00210-003-0779-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2003] [Accepted: 06/12/2003] [Indexed: 10/26/2022]
Abstract
Magnolol, an active component extracted from Magnolia officinalis, has various pharmacological effects, including potent antioxidant activity. In the present study, we investigated the effect of magnolol on apoptosis in rat vascular smooth muscle cells (VSMCs), using terminal-deoxynucleotidyl-transferase-mediated deoxyuridine triphosphate nick end labelling (TUNEL) and flow cytometric analysis. Magnolol (5-20 micro M) concentration-dependently induced significant VSMC apoptosis, this effect being blocked by the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (z-VAD-fmk, 50 micro M). To study the molecular mechanism, the mitochondrial death pathway was examined. Magnolol increased caspase-3 and caspase-9 activities significantly and reduced the mitochondrial potential (Deltapsi(m)). The levels of B-cell leukaemia/lymphoma-2 (Bcl-2), but not those of Bcl-2-associated X protein (Bax) or Bcl-x(L), were down-regulated concentration dependently by magnolol. In an animal model, balloon angioplasty-induced neointima formation was inhibited significantly by magnolol and Bcl-2 protein levels were reduced. Taken together, these results show that magnolol induces apoptosis in VSMCs via the mitochondrial death pathway. This effect is mediated through down-regulation of Bcl-2 protein levels, both in vivo and in vitro. Magnolol thus shows potential as a novel therapeutic agent for the treatment of atherosclerosis and re-stenosis.
Collapse
MESH Headings
- Angioplasty, Balloon
- Animals
- Antioxidants/pharmacology
- Apoptosis/drug effects
- Biphenyl Compounds/pharmacology
- Carotid Artery Diseases/etiology
- Carotid Artery Diseases/pathology
- Caspase 3
- Caspase 9
- Caspases/biosynthesis
- Cells, Cultured
- Constriction, Pathologic
- Down-Regulation
- Enzyme Activation
- Lignans
- Male
- Membrane Potentials/drug effects
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins c-bcl-2/biosynthesis
- Rats
- Rats, Sprague-Dawley
- bcl-2-Associated X Protein
- bcl-X Protein
Collapse
Affiliation(s)
- Jiun-Han Chen
- National Defence Medical Centre, 114, Taipei, Taiwan
| | | | | | | |
Collapse
|
31
|
Hutter R, Sauter BV, Reis ED, Roque M, Vorchheimer D, Carrick FE, Fallon JT, Fuster V, Badimon JJ. Decreased reendothelialization and increased neointima formation with endostatin overexpression in a mouse model of arterial injury. Circulation 2003; 107:1658-63. [PMID: 12668502 DOI: 10.1161/01.cir.0000058169.21850.ce] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Impaired endothelial regeneration contributes to arterial lesion formation. Endostatin is a specific inhibitor of endothelial cell growth and induces endothelial cell apoptosis. We examined the effect of endostatin overexpression on reendothelialization and neointima formation in a mouse model of arterial injury. METHODS AND RESULTS Mice underwent femoral arterial denudation and received recombinant adenovirus, expressing either murine endostatin (n=19) or control adenoviral vector (n=12), by jugular vein injection. Endostatin gene transfer resulted in high serum levels of endostatin. Strong adenoviral gene expression of beta-galactosidase-expressing control vector was detected in liver tissue and was absent in the injured arterial wall at 1 week. Deposits of endostatin protein were detected along the denuded arterial wall and were not seen in the noninjured contralateral artery at 1 week. Endostatin deposits were also absent in the injured artery of control vector-treated animals. Overexpression of endostatin led to decreased reendothelialization and increased apoptosis of luminal endothelial cells 2 and 4 weeks after arterial injury (P<0.05). In addition, endostatin overexpression resulted in increased neointima formation (P<0.05). Endothelial apoptosis and neointima area correlated positively with endostatin serum levels, whereas the degree of reendothelialization correlated negatively with endostatin serum levels (P<0.05). Furthermore, poor reendothelialization correlated with increased neointima formation (P<0.05). CONCLUSIONS In summary, decreased reendothelialization and enhanced endothelial apoptosis, in response to endostatin overexpression, were associated with increased neointima formation. These findings demonstrate that high serum levels of endostatin are capable of inhibiting endothelial regeneration and promoting arterial lesion growth in conditions of endothelial injury.
Collapse
Affiliation(s)
- Randolph Hutter
- Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, 1425 Madison Ave, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Braun-Dullaeus RC, Ziegler A, Bohle RM, Bauer E, Hein S, Tillmanns H, Haberbosch W. Quantification of the cell-cycle inhibitors p27(Kip1) and p21(Cip1) in human atherectomy specimens: primary stenosis versus restenosis. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2003; 141:179-89. [PMID: 12624599 DOI: 10.1067/mlc.2003.23] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proliferation, a key determinator of vascular proliferative diseases, is dependent on cyclin/cyclin-dependent kinase (CDK) complexes, which are controlled by cyclin-dependent kinase inhibitors (CKIs) such as p27(Kip1) and p21(Cip1). Both have prognostic significance in various human malignancies. We have determined the levels of p27(Kip1) and p21(Cip1) in human directional coronary atherectomy specimens of primary lesions (n = 15) and lesions of in-stent restenosis (n = 18) in comparison to those of other vascular regions and have correlated CKI levels with clinical data. Quantitative immunoblotting demonstrated low expression of p27(Kip1) in primary lesions (5.9 +/- 0.5 ng/mg protein) compared with that in aorta (14.9 +/- 0.9 ng/mg), internal mammary artery (16.7 +/- 1.1 ng/mg), and carotid artery thrombendarterectomy specimens (16.5 +/- 1.7 ng/mg). Similarly, p27(Kip1) levels in lesions of in-stent restenosis were found to be significantly reduced (6.3 +/- 1.1 ng/mg; mean time of restenosis development 367 +/- 61 days). p27(Kip1) levels did, however, not have prognostic significance for the development of restenosis, and expression levels of proliferating cell nuclear antigen and CDK2 were similar in all groups examined, indicating low proliferative activity. Clinically, p27(Kip1) was not of value in predicting the development of restenosis. Furthermore, p27(Kip1) tissue levels were not increased in statin-treated patients, implying that the favorable effect of these drugs is not a result of p27(Kip1) stabilization. However, the relative content of p21(Cip1) was found to be significantly up-regulated in restenosis compared with that in primary lesions (225%) and the other vascular regions. Our data imply that negative-feedback mechanisms are still intact in coronary proliferative disease, thereby contrasting the finding of deregulated proliferation in malignancies.
Collapse
Affiliation(s)
- Ruediger C Braun-Dullaeus
- Department of Internal Medicine, Giessen University, the Kerckhoff Klinik, Bad Nauheim, and Zentralklinikum Suhl, Germany.
| | | | | | | | | | | | | |
Collapse
|
33
|
Chung IM, Gold HK, Schwartz SM, Ikari Y, Reidy MA, Wight TN. Enhanced extracellular matrix accumulation in restenosis of coronary arteries after stent deployment. J Am Coll Cardiol 2002; 40:2072-81. [PMID: 12505216 DOI: 10.1016/s0735-1097(02)02598-6] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVES The goal of this study was to evaluate the cellular and extracellular composition of human coronary arterial in-stent restenosis after various periods of time following stent deployment. BACKGROUND Neointimal in-growth rather than stent recoil is thought to be important for coronary arterial in-stent restenosis. There is only limited data on the cellular and extracellular composition changes with time after stent deployment. METHODS We analyzed 29 coronary arterial in-stent restenotic tissue samples (14 left anterior descending coronary artery, 10 right coronary artery, and 5 left circumflex artery) retrieved by using directional coronary atherectomy from 25 patients at 0.5 to 23 (mean, 5.7) months after deployment of Palmaz-Schatz stents employing histochemical and immunocytochemical techniques. RESULTS Cell proliferation was low (0% to 4%). Myxoid tissue containing extracellular matrix (ECM) enriched with proteoglycans was found in 69% of cases and decreased over time after stenting. Cell-depleted areas were found in 57% of cases and increased with time after stenting. Versican, biglycan, perlecan, and hyaluronan were present with varying individual distributions in all samples. Positive transforming growth factor-beta1 staining was found in 80% of cases. Immunostaining with alpha-smooth muscle actin identified the majority of cells as smooth muscle cells with occasional macrophages present (< or =12 cells per section). CONCLUSIONS These data suggest that enhanced ECM accumulation rather than cell proliferation contribute to later stages of in-stent restenosis. Balloon angioplasty of in-stent restenosis may, therefore, fail due to ECM changes during: 1) additional stent expansion, 2) tissue extrusion out of the stent, or 3) tissue compression.
Collapse
Affiliation(s)
- Ick-Mo Chung
- Department of Pathology, University of Washington, Seattle, USA
| | | | | | | | | | | |
Collapse
|
34
|
Glover C, Ma X, Chen YX, Miller H, Veinot J, Labinaz M, O'Brien E. Human in-stent restenosis tissue obtained by means of coronary atherectomy consists of an abundant proteoglycan matrix with a paucity of cell proliferation. Am Heart J 2002; 144:702-9. [PMID: 12360168 DOI: 10.1067/mhj.2002.123577] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND In-stent restenosis (ISR) is a shortcoming of percutaneous coronary revascularization. Although neointimal cell proliferation is suspected to be the cause of this problem, little histological characterization of this tissue or data on cell replication exist. The purpose of this study was to examine the histologic features and proliferation profile of coronary ISR tissue derived from atherectomy procedures performed on patients with clinical evidence of ISR. METHODS ISR tissue retrieved by means of atherectomy from 20 coronary lesions was subjected to histomorphological analyses and immunocytochemistry as a means of examining proteoglycan expression. Cell proliferation was assessed with 2 sensitive markers of replication, in situ hybridization for histone 3 messenger RNA expression and immunocytochemistry for Ki-67 expression. RESULTS The ISR atherectomy specimens consisted primarily of smooth muscle cells, with occasional focal collections of inflammatory cells and organizing thrombus. All specimens had low levels of interstitial collagen and an abundant proteoglycan matrix, with biglycan being overexpressed more commonly than decorin. Cell proliferation was found in only 1 of 20 specimens (2 cells). CONCLUSION Established ISR lesions contained an abundant proteoglycan matrix and a paucity of proliferating cells. Future therapeutic strategies for ISR should include targeting extracellular matrix production.
Collapse
Affiliation(s)
- Chris Glover
- Vascular Biology Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
35
|
Peppel K, Zhang L, Huynh TTT, Huang X, Jacobson A, Brian L, Exum ST, Hagen PO, Freedman NJ. Overexpression of G protein-coupled receptor kinase-2 in smooth muscle cells reduces neointimal hyperplasia. J Mol Cell Cardiol 2002; 34:1399-1409. [PMID: 12393000 DOI: 10.1006/jmcc.2002.2092] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The activation of vascular smooth muscle cells (SMCs) in neointimal hyperplasia involves signaling through receptor tyrosine kinases as well as G protein-coupled receptors. Overexpression of G protein-coupled receptor kinase-2 (GRK2) in SMCs can attenuate mitogenic signaling and proliferation in response to not only several G protein-coupled receptor agonists, but also platelet-derived growth factor (PDGF). To test whether overexpression of GRK2 could inhibit other SMC responses implicated in neointimal hyperplasia, we assessed SMC chemotaxis and mitogenic signaling evoked by PDGF and G(q)-coupled receptor agonists. To test the effects of GRK2 overexpression on neointimal hyperplasia in vivo, we employed a rabbit autologous vein graft model system. GRK2 overexpression reduced PDGF-promoted SMC chemotaxis by 85% (P<0.01), but had no effect on chemotaxis promoted by epidermal growth factor (EGF). Congruently, GRK2 overexpression reduced by approximately 50% (P<0.05) the [(3)H]thymidine incorporation induced by combinations of PDGF and Gq-coupled receptor agonists, but had no effect on that induced by PDGF plus EGF. PDGF-, but not EGF-promoted phosphoinositide 3-kinase activity in SMCs was also inhibited by GRK2 overexpression. In rabbit vein grafts, we achieved GRK2 overexpression in medial SMCs, reduced cell proliferation during the first week after graft implantation, and reduced steady state neointimal thickness by 29% (P<0.01), without affecting medial thickness or potentiating SMC apoptosis. Because of its ability to dampen chemotactic and mitogenic signaling through PDGF and Gq-coupled receptors, GRK2 overexpression in SMCs may be a useful therapeutic approach for neointimal hyperplasia.
Collapse
Affiliation(s)
- Karsten Peppel
- Duke University Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hilker M, Tellmann G, Buerke M, Gloger K, Moersig W, Oelert H, Hake U, Lehr HA. Proliferative activity in stenotic human aortocoronary bypass grafts. Cardiovasc Pathol 2002; 11:284-90. [PMID: 12361839 DOI: 10.1016/s1054-8807(02)00113-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Aortocoronary bypass graft disease is responsible for long-term failure of autologous vein grafts. The analyses of proliferation and cell type characterisation in human bypass grafts harvested during re-do surgery make it possible to investigate the cellular processes leading to bypass graft failure. METHODS 30 stenotic vein grafts and 25 control veins were explantated during re-do heart surgery procedures. The total area and cell count of the neointima, media and adventitia were calculated computer-assisted. Actively proliferating cells were identified using antibody to Ki-67 and positive cells were determined by double-label immunocytochemistry with SMC alpha-actin, CD 31 (endothelial cells), CD 68 (macrophages) and CD 45 (T-lymphocytes). RESULTS Active proliferation was detected in different cell types with an average proliferation index of 0.15%, 0.18% and 0.086% for neointima, media and adventitia. Only 9% of proliferating cells in the neointima were SMC (not identified cells 40%); correspondingly, 14% SMC (not identified cells 33%) were detected in the media. Endothelial cells turned out to be the predominant proliferating cell type in all sections of the vessel wall. CONCLUSION Proliferation in our series of stenotic vein grafts occurred at a low level, but was significantly higher compared to native control veins. While proliferation may play an important role in early lesions, our data clearly show low proliferation activity in advanced graft lesions. The identification of proliferating macrophages and T-lymphocytes implicate an additional inflammatory component in the development of human bypass graft disease. SUMMARY To clarify the role of cellular proliferation in human aortocoronary bypass grafts, we characterized the cellular composition and proliferation index in 30 stenotic saphenous vein grafts in comparison to 25 native veins. Proliferation in our series of stenotic vein grafts occurred at a low level, but was significantly higher compared to native control veins.
Collapse
Affiliation(s)
- Michael Hilker
- Department of Thoracic and Cardiovascular Surgery, Johannes Gutenberg University Mainz, Langenbeckstr. 1, D-55131 Mainz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Sakuma H, Yamamoto M, Okumura M, Kojima T, Maruyama T, Yasuda K. High glucose inhibits apoptosis in human coronary artery smooth muscle cells by increasing bcl-xL and bfl-1/A1. Am J Physiol Cell Physiol 2002; 283:C422-8. [PMID: 12107051 DOI: 10.1152/ajpcell.00577.2001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is a serious complication in diabetic patients. To elucidate the precise mechanisms of atherosclerosis in diabetic patients, the effects of high glucose concentration (25 mM) on apoptosis regulation and bcl-2 family protein expression in human coronary artery smooth muscle cells (CASMC) were examined. Treatment with a high level of glucose (25 mM) caused a significant decrease in apoptosis in CASMC compared with the same cells treated with a physiologically normal glucose concentration (5.5 mM) (23.9 +/- 2.4% vs. 16.5 +/- 1.8%; P < 0.01). With respect to apoptosis regulation, treatment of CASMC with high glucose concentration markedly increased mRNA expressions of bcl-xL and bfl-1/A1 compared with cells treated with normal glucose. High glucose induced phosphorylation of phosphatidylinositol 3-kinase (PI 3-K) and extracellular signal-regulated kinase (ERK)1/2 along with bcl-xL and bfl-1/A1 upregulation. These results suggest that high glucose suppresses apoptosis via upregulation of bcl-xL and bfl-1/A1 levels through PI 3-K and ERK1/2 pathways in CASMC. High glucose-induced increase in the expression of antiapoptotic proteins may be important in the development of atherosclerosis in diabetic patients.
Collapse
Affiliation(s)
- Hiroya Sakuma
- Third Department of Internal Medicine, Gifu University School of Medicine, Gifu 500-8705, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Within the past decade, focus on radiation to prevent restenosis has moved from a concept developed in the animal laboratory to a clinical treatment. The initial evaluation of coronary artery radiation therapy focused on changes in the function of the artery or lesion formation following overstretch balloon injury in pigs. A number of concepts emerged from this work: (1) radiation inhibits neointima formation in a dose-dependent fashion, (2) radiation prevents negative remodeling, (3) radiation does not reverse established injury, (4) low dose irradiation in an injured area may be injurious, (5) radiation is a useful adjunct to stenting, (6) benefits of radiation in animal models at 6 months are less pronounced than at 1 month, (7) radiation delays healing, (8) permanent stents and radiation delivered from external sources may have very different effects on restenosis, and (9) radiation interferes with vessel wall function. More recent studies of irradiation have looked at the molecular biological effects of radiation in hopes of understanding how this therapy works, and how it may be improved. This article attempts to summarize the known animal and cellular work on radiation in preventing restenosis.
Collapse
Affiliation(s)
- Ian Crocker
- Emory University School of Medicine, and Atlanta Cardiovascular Research Institute, Atlanta, GA, USA.
| | | |
Collapse
|
39
|
Wang Y, Shiota N, Leskinen MJ, Lindstedt KA, Kovanen PT. Mast cell chymase inhibits smooth muscle cell growth and collagen expression in vitro: transforming growth factor-beta1-dependent and -independent effects. Arterioscler Thromb Vasc Biol 2001; 21:1928-33. [PMID: 11742866 DOI: 10.1161/hq1201.100227] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the vulnerable areas of fibrous caps of advanced atherosclerotic lesions, chymase-containing mast cells are present. In such areas, the numbers of smooth muscle cells (SMCs) and the content of collagen are reduced. In this in vitro study, we found that the addition of chymase, isolated and purified from rat serosal mast cells, to cultured rat aortic SMCs of the synthetic phenotype (s-SMCs) inhibited their proliferation by blocking the G(0)/G(1)-->S transition in the cell cycle. Rat chymase and recombinant human chymase inhibited the expression of collagen type I and type III mRNA in s-SMCs and in human coronary arterial SMCs. The growth-inhibitory effect of chymase was partially reversed by addition to the culture medium of an antibody capable of neutralizing the activity of transforming growth factor-beta1 (TGF-beta1). Immunocytochemistry showed that the s-SMCs expressed and synthesized extracellular matrix-associated TGF-beta1. On exposure to mast cell chymase, the extracellular matrix-associated latent TGF-beta1 was released and activated, as demonstrated by immunoblotting and by an ELISA with TGF-beta1 type II receptor for capture. When added to s-SMCs, such chymase-released TGF-beta1 was capable of inhibiting their growth. In contrast, the inhibitory effect of chymase on collagen synthesis by s-SMCs did not depend on TGF-beta1. Taken together, the findings support the hypothesis that chymase released from activated mast cells in atherosclerotic plaques contributes to cap remodeling.
Collapse
Affiliation(s)
- Y Wang
- Wihuri Research Institute, Helsinki, Finland
| | | | | | | | | |
Collapse
|
40
|
Chaulet H, Desgranges C, Renault MA, Dupuch F, Ezan G, Peiretti F, Loirand G, Pacaud P, Gadeau AP. Extracellular nucleotides induce arterial smooth muscle cell migration via osteopontin. Circ Res 2001; 89:772-8. [PMID: 11679406 DOI: 10.1161/hh2101.098617] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Migration and proliferation of arterial smooth muscle cells (SMCs) play a prominent role in the development of atherosclerotic plaques and restenosis lesions. Most of the growth-regulatory molecules potentially involved in these pathological conditions also demonstrate chemotactic properties. Extracellular purine and pyrimidine nucleotides have been shown to induce cell cycle progression and to elicit growth of cultured vascular SMCs. Moreover, the P2Y(2) ATP/UTP receptor was overexpressed in intimal thickening, suggesting a role of these nucleotides in vascular remodeling. Using the Transwell system migration assay, we demonstrate that extracellular ATP, UTP, and UDP exhibit a concentration-dependent chemotactic effect on cultured rat aortic SMCs. UTP, the most powerful nucleotide inducer of migration, elicited significant responses from 10 nmol/L. In parallel, UTP increased osteopontin expression dose-dependently. The blockade of osteopontin or its integrin receptors alpha(v)beta(3)/beta(5) by specific antibodies or antagonists inhibited UTP-induced migration. Moreover, the blockade of ERK-1/ERK-2 MAP kinase or rho protein pathways led to the inhibition of both UTP-induced osteopontin increase and migration, demonstrating the central role of osteopontin in this process. Taken together, these results suggest that extracellular nucleotides, and particularly UTP, can induce arterial SMC migration via the action of osteopontin.
Collapse
MESH Headings
- Adenosine Diphosphate/pharmacology
- Adenosine Triphosphate/pharmacology
- Animals
- Aorta
- Calcium/metabolism
- Cell Movement/drug effects
- Cell Movement/physiology
- Cells, Cultured
- Chemotaxis/drug effects
- Chemotaxis/physiology
- Diffusion Chambers, Culture
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Extracellular Space/metabolism
- Intracellular Signaling Peptides and Proteins
- Mitogen-Activated Protein Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Nucleotides/pharmacology
- Oligopeptides/pharmacology
- Osteopontin
- Phosphorylation/drug effects
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Rats
- Rats, Wistar
- Sialoglycoproteins/metabolism
- Uridine Diphosphate/pharmacology
- Uridine Triphosphate/pharmacology
- rho-Associated Kinases
- rhoA GTP-Binding Protein/metabolism
Collapse
|
41
|
Bennett MR, O'Sullivan M. Mechanisms of angioplasty and stent restenosis: implications for design of rational therapy. Pharmacol Ther 2001; 91:149-66. [PMID: 11728607 DOI: 10.1016/s0163-7258(01)00153-x] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Restenosis after angioplasty or stenting remains the major limitation of both procedures. A vast array of drug therapies has been used to prevent restenosis, but they have proven to be predominantly unsuccessful. Recent trends in drug therapy have attempted to refine the molecular and biological targets of therapy, based on the assumption that a single biological process or molecule is critical to restenosis. In contrast, both stenting and brachytherapy, which are highly nonspecific, can successfully reduce restenosis after angioplasty or stenting, respectively. This review examines the biology of both angioplasty and stent stenosis, focussing on human studies. We also review the landmark human trials that have definitively proven successful therapies, such as stenting and brachytherapy. We suggest that the successful trials of stenting and brachytherapy and the failure of other treatments have highlighted the shortcomings of conventional animal models of arterial intervention, and gaps in our knowledge of human disease. In contrast to arguments advocating gene therapy, these studies suggest that the most likely successful drug therapy will have a wide therapeutic range, targeting as many of the components or biological processes contributing to restenosis as possible.
Collapse
Affiliation(s)
- M R Bennett
- Division of Cardiovascular Medicine, Addenbrooke's Centre for Clinical Investigation, Box 110, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK.
| | | |
Collapse
|
42
|
Murakami S, Toda Y, Seki T, Munetomo E, Kondo Y, Sakurai T, Furukawa Y, Matsuyama M, Nagate T, Hosokawa N, Nagata K. Heat shock protein (HSP) 47 and collagen are upregulated during neointimal formation in the balloon-injured rat carotid artery. Atherosclerosis 2001; 157:361-8. [PMID: 11472735 DOI: 10.1016/s0021-9150(00)00743-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Heat shock protein (HSP) 47, a collagen-specific molecular chaperone, is thought to be essential for the proper processing and secretion of procollagen molecules. We investigated the time course and localization of HSP47 and collagen expression after balloon catheter angioplasty in the rat carotid artery, based on the premise that accumulation of extracellular matrix components is a main feature of intimal hyperplasia in humans and in laboratory animals. Low levels of HSP47 expression were evident in uninjured carotid arteries. Northern blot analysis revealed that HSP47 mRNA expression was markedly stimulated 1--3 days after the induced injury and a high level was maintained for 7 days, followed by a gradual decline for up to 21 days after the injury. These changes in HSP47 expression paralleled changes in alpha 1(I) collagen expression. Immunohistochemical staining revealed colocalization of HSP47 and collagen in smooth muscle cells (SMCs) of the media and intima. In situ hybridization analysis showed that activated SMCs, which proliferated and migrated into the intima, expressed high levels of HSP47. In cultured human aortic SMCs, similar upregulation of HSP47 and alpha1(I) collagen by TGF-beta was noted. These results show that SMCs activated after balloon injury express high levels of HSP47 and collagen during cell proliferation and migration, hence an overproduction of collagen and development of intimal thickening. Thus, HSP47 plays a role in the formation and progression of neointima after angioplasty.
Collapse
Affiliation(s)
- S Murakami
- Medicinal Research Laboratories, Taisho Pharmaceutical Co. Ltd., Ohmiya 330-8530, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Schoenhagen P, Ziada KM, Vince DG, Nissen SE, Tuzcu EM. Arterial remodeling and coronary artery disease: the concept of "dilated" versus "obstructive" coronary atherosclerosis. J Am Coll Cardiol 2001; 38:297-306. [PMID: 11499716 DOI: 10.1016/s0735-1097(01)01374-2] [Citation(s) in RCA: 190] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Traditionally, the development of coronary artery disease (CAD) was described as a gradual growth of plaques within the intima of the vessel. The outer boundaries of the intima, the media and the external elastic membrane (EEM), were thought to be fixed in size. In this model plaque growth would always lead to luminal narrowing and the number and severity of angiographic stenoses would reflect the extent of coronary disease. However, histologic studies demonstrated that certain plaques do not reduce luminal size, presumably because of expansion of the media and EEM during atheroma development. This phenomenon of "arterial remodeling" was confirmed in necropsy specimens of human coronary arteries. More recently, the development of contemporary imaging technology, particularly intravascular ultrasound, has allowed the study of arterial remodeling in vivo. These new imaging modalities have confirmed that plaque progression and regression are not closely related to luminal size. In this review, we will analyze the role of remodeling in the progression and regression of native CAD, as well as its impact on restenosis after coronary intervention.
Collapse
|
44
|
McLaughlin R, Kelly CJ, Kay E, Bouchier-Hayes D. The role of apoptotic cell death in cardiovascular disease. Ir J Med Sci 2001; 170:132-40. [PMID: 11491050 PMCID: PMC7102203 DOI: 10.1007/bf03168827] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Programmed cell death, or apoptosis, is a distinct, managed form of cell death. It is fundamentally different from necrosis. It is a genetically controlled, energy-dependent method of cellular deletion without inflammation. In the cardiovascular system, apoptosis occurs as a primary and secondary event in disease pathogenesis. This review addresses our current understanding of the initiation, propagation and significance of apoptosis in the cardiovascular system, as well as assessing therapeutic potentials arising therefrom. METHODS A Medline search was performed and relevant publications reviewed. Further articles were obtained from the references of these publications. RESULTS and conclusions Apoptotic cell death is a key element in the pathogenesis and progression of ischaemia-reperfusion (IR) injury, cardiac failure, myocardial infarction, atherosclerosis, endothelial dysfunction and the clinical syndromes which these situations produce. Our increased understanding of the role of apoptosis in the pathogenesis of cardiovascular disease offers potential to develop new therapeutic strategies.
Collapse
Affiliation(s)
- R McLaughlin
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | | | | | | |
Collapse
|
45
|
Kiesz RS, Buszman P, Martin JL, Deutsch E, Rozek MM, Gaszewska E, Rewicki M, Seweryniak P, Kosmider M, Tendera M. Local delivery of enoxaparin to decrease restenosis after stenting: results of initial multicenter trial: Polish-American Local Lovenox NIR Assessment study (The POLONIA study). Circulation 2001; 103:26-31. [PMID: 11136681 DOI: 10.1161/01.cir.103.1.26] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Enoxaparin inhibits smooth muscle cell proliferation in experimental models. Intimal hyperplasia has been found to be the principal cause of restenosis after coronary stent implantation. We sought to determine whether the intramural delivery of enoxaparin before stenting of de novo lesions decreases restenosis. METHODS AND RESULTS One hundred patients who were undergoing stenting were randomly assigned to either local administration of enoxaparin during predilation with reduced systemic heparinization or stenting with standard, systemic heparinization. All patients were treated with the same type of stent (NIR). The primary study end point was late luminal loss. The secondary end points were major adverse cardiac events, target lesion revascularization, and angiographic restenosis at 6 months. Angiographic follow-up at 6 months was completed in all except 1 patient. Late luminal loss was reduced to 0.76+/-0.42 mm in the local enoxaparin delivery group versus 1. 07+/-0.49 mm in the systemic heparinization group (P:<0.001). Restenosis, using a binary definition, occurred in 10% of patients in the enoxaparin group and in 24% of patients in the systemic heparinization group (P:<0.05). Target lesion revascularization rates occurred in 8% of the enoxaparin group and 22% of the systemic heparinization group (P:<0.05). There were no deaths and no emergent CABGs were performed. The only subacute stent closure and non-Q-wave infarction occurred in a patient assigned to the systemic heparinization group. CONCLUSIONS This is the first prospective randomized trial in which the local delivery of a drug, enoxaparin, resulted in significant reduction in late luminal loss and restenosis after stent implantation in de novo coronary lesions.
Collapse
Affiliation(s)
- R S Kiesz
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, TX 78229-7872, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Affiliation(s)
- Ilia Fishbein
- School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael Chorny
- School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gershon Golomb
- School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
47
|
O'Brien ER, Urieli-Shoval S, Garvin MR, Stewart DK, Hinohara T, Simpson JB, Benditt EP, Schwartz SM. Replication in restenotic atherectomy tissue. Atherosclerosis 2000; 152:117-26. [PMID: 10996346 DOI: 10.1016/s0021-9150(99)00457-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Previously, we demonstrated that replication in restenotic coronary atherectomy specimens was an infrequent and modest event. In general, this data was interpreted with caution, as immunocytochemistry for the proliferating cell nuclear antigen (PCNA) was used to subjectively assess proliferation and most of the tissue specimens were resected more than 3 months after the initial interventional procedure. The purpose of the present study was to use a more sensitive method of detecting replication, in situ hybridization for histone 3 (H3) mRNA, to determine the replication profile of human directional atherectomy specimens. Restenotic directional coronary atherectomy specimens from lesions that had undergone an interventional procedure within the preceding 3 months were studied. In addition, larger atherectomy specimens from peripheral arterial lesions were assessed to ensure that pockets of replication were not being overlooked in the smaller coronary specimens. We found evidence for replication in tissue resected from 2/17 coronary and 9/12 peripheral artery restenotic lesions. In contrast, 3/11 specimens resected from primary lesions of peripheral arteries also expressed H3 mRNA. We estimated that the maximum percentage of cells that were replicating in restenotic coronary, restenotic peripheral and primary peripheral artery tissue slides to be <0.5, < or =1.2 and <0.01%, respectively. Replication was found in tissue specimens resected both early and late after a previous interventional procedure. For specimens with >15 replicating cells per slide we found high levels of focal replication. Therefore, cell replication, as assessed by the expression of H3 mRNA, was infrequent in restenotic coronary artery specimens, whereas peripheral restenotic lesions had more frequent and higher levels of replication regardless of the interval from the previous interventional procedure. For all specimens the percentage of cells that were replicating was low, however focal areas with relatively high replication indices were presented. Although replication was more abundant in restenotic lesions it does not appear to be a dominant event in the pathophysiology of restenosis.
Collapse
Affiliation(s)
- E R O'Brien
- Vascular Biology Laboratory, Department of Medicine (Cardiology), University of Ottawa Heart Institute, 40 Ruskin Street, Ont., K1Y 4W7, Ottawa, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Consigny PM, Kwak A, Vitali NJ. Immunolocalization of proliferating cells in the rabbit iliac artery after balloon angioplasty. J Vasc Interv Radiol 2000; 11:206-12. [PMID: 10716391 DOI: 10.1016/s1051-0443(07)61466-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Experiments were performed to characterize the location of proliferating cells in the balloon-dilated rabbit iliac artery. MATERIALS AND METHODS Balloon angioplasty was performed on the external iliac arteries in each of four rabbits. The arteries were removed 3 days later, frozen, cryosectioned, and immunostained with Ki-67, an antibody that identifies proliferating cells. The sections were then examined to determine the patterns of cell proliferation within the arterial media and the ratio of proliferating to nonproliferating cells. RESULTS Of the 31 arterial cross-sections examined, cell proliferation was circumferential in five (16%), and focal in 26 (84%). Of the 86 foci of proliferation examined within the 31 cross-sections, proliferation was localized to the inner media in 30 (35%), to the outer media in four (5%), and was transmural in 52 (60%). The internal elastica lamina (IEL) appeared normal at 22 foci (26%), but appeared stretched or torn at 64 (74%). Proliferation was usually confined to the inner media at foci having no IEL injury (18 of 22; 82%), but was most often transmural where the IEL was stretched or torn (49 of 64; 77%). The ratio of proliferating to nonproliferating cells, which averaged 0.31 +/- .20, was greater (P < .01) in areas with IEL injury than in areas without IEL injury. CONCLUSION These results suggest that angioplasty-induced cell proliferation is typically focal rather than circumferential and is associated with stretching or tearing of the IEL.
Collapse
Affiliation(s)
- P M Consigny
- Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|
49
|
Abstract
Complexing recombinant DNA with cationic liposomes is a convenient means of introducing foreign genes into cells (lipofection) and could potentially form the basis for genetically modifying diseased blood vessels in patients. The mechanism of lipofection is incompletely understood, but it is recognized that the degree of successful gene transfer is highly dependent on cell type. We have transfected primary cultures of human vascular smooth muscle cells with a plasmid expressing either firefly luciferase (Luc) or nuclear-localized beta-galactosidase (NL-beta-gal). Cells were derived from either normal human internal mammary arteries, fragments of primary atherosclerotic plaque, or fragments of restenotic lesion. Concurrent lipofection of rabbit vascular smooth muscle cells and NIH 3T3 cells was performed as well. Compared with NIH 3T3 cells, expression in human vascular smooth muscle cells was markedly reduced: In cells derived from internal mammary artery, Luc expression, normalized for protein content, was 123-fold lower than in NIH 3T3 cells, while the proportion of cells expressing NL-beta-gal was 30-fold lower. Luc expression in cells derived from restenotic tissue was significantly greater than from cells derived from primary plaque. Within a given population of cells, the mitotic index of cells expressing the recombinant gene was significantly higher than the mitotic index for the total population of cells (p < 0.05). Finally, cotransfection experiments, in which lipofection of smooth muscle cells was performed using genes for NL-beta-gal and for human growth hormone, showed that among positive transfectants a high proportion of cells (23-36%) coexpressed both genes. Thus, the efficiency of successful lipofection in human vascular smooth muscle cells in vitro is low. Transfection appears to be preferentially facilitated in cells derived from restenotic tissue and specific properties of smooth muscle cells, including growth rates, appear to be critical for successful transfection. Further elucidation of cell properties that promote transfection is required to augment the efficiency of liposome-mediated gene transfer in human vascular cells.
Collapse
|
50
|
Bortman SM, Losordo DW. Dynamics of Vascular Remodeling: An Overview and Bibliography. J Thromb Thrombolysis 1999; 3:71-86. [PMID: 10608041 DOI: 10.1007/bf00226415] [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: 11/24/2022]
Affiliation(s)
- SM Bortman
- Department of Cardiology, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts and Department of Cardiovascular Research, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | | |
Collapse
|