1
|
Lin HF, Wu MN, Chen CY, Lim K, Juo SHH, Chen CS. Thrombospondin-1 associated with carotid intima-media thickness among individuals with hypertension. J Investig Med 2024; 72:279-286. [PMID: 38217383 DOI: 10.1177/10815589241228589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
In vivo and in vitro studies have demonstrated that thrombospondin-1 (TSP-1) is involved in atherosclerotic pathogenesis. However, the role of TSP-1 in clinical atherosclerosis remains unknown. This cross-sectional study investigated the relationship between TSP-1 and carotid intima-media thickness (IMT) and examined whether it interacts with conventional cardiovascular risk factors. A total of 587 participants were enrolled from February 2018 to December 2021. TSP-1 was dichotomized based on median value. Carotid IMT was measured bilaterally in each segment, and the average value was taken as the overall IMT variable. Analysis of covariance models were used to ascertain the main and interaction effects of cardiovascular risk factors and circulating TSP-1 levels on carotid IMT. Those with high TSP-1 (n = 294) had significantly higher carotid IMT than did those with low TSP-1 (n = 293; 0.74 ± 0.12 vs 0.72 ± 0.11 mm; p = 0.011). After the combined effects of TSP-1 and vascular risk factors on carotid IMT were evaluated, an interaction effect on IMT was observed between TSP-1 and hypertension (adjusted F = 8.760; p = 0.003). Stratification analysis revealed that individuals with hypertension and high TSP-1 had significantly higher IMT than did those with low TSP-1 (adjusted p = 0.007). However, this difference was not observed in normotensive individuals (adjusted p = 0.636). In conclusion, this is the first study to provide clinical data supporting the correlation between TSP-1 and atherosclerosis. TSP-1 may be a crucial marker of increased susceptibility to atherosclerosis in individuals with hypertension.
Collapse
Affiliation(s)
- Hsiu-Fen Lin
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Meng-Ni Wu
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Yuan Chen
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kelly Lim
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Suh-Hang Hank Juo
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan
- Drug Development Center, China Medical University, Taichung, Taiwan
| | - Cheng-Sheng Chen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Psychiatry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
2
|
McQueen A, Escuer J, Schmidt AF, Aggarwal A, Kennedy S, McCormick C, Oldroyd K, McGinty S. An intricate interplay between stent drug dose and release rate dictates arterial restenosis. J Control Release 2022; 349:992-1008. [PMID: 35921913 DOI: 10.1016/j.jconrel.2022.07.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/14/2022] [Accepted: 07/26/2022] [Indexed: 10/15/2022]
Abstract
Since the introduction of percutaneous coronary intervention (PCI) for the treatment of obstructive coronary artery disease (CAD), patient outcomes have progressively improved. Drug eluting stents (DES) that employ anti-proliferative drugs to limit excess tissue growth following stent deployment have proved revolutionary. However, restenosis and a need for repeat revascularisation still occurs after DES use. Over the last few years, computational models have emerged that detail restenosis following the deployment of a bare metal stent (BMS), focusing primarily on contributions from mechanics and fluid dynamics. However, none of the existing models adequately account for spatiotemporal delivery of drug and the influence of this on the cellular processes that drive restenosis. In an attempt to fill this void, a novel continuum restenosis model coupled with spatiotemporal drug delivery is presented. Our results indicate that the severity and time-course of restenosis is critically dependent on the drug delivery strategy. Specifically, we uncover an intricate interplay between initial drug loading, drug release rate and restenosis, indicating that it is not sufficient to simply ramp-up the drug dose or prolong the time course of drug release to improve stent efficacy. Our model also shows that the level of stent over-expansion and stent design features, such as inter-strut spacing and strut thickness, influence restenosis development, in agreement with trends observed in experimental and clinical studies. Moreover, other critical aspects of the model which dictate restenosis, including the drug binding site density are investigated, where comparisons are made between approaches which assume this to be either constant or proportional to the number of smooth muscle cells (SMCs). Taken together, our results highlight the necessity of incorporating these aspects of drug delivery in the pursuit of optimal DES design.
Collapse
Affiliation(s)
- Alistair McQueen
- Division of Biomedical Engineering, University of Glasgow, Glasgow, UK
| | - Javier Escuer
- Aragón Institute for Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | | | - Ankush Aggarwal
- Glasgow Computational Engineering Centre, Division of Infrastructure and Environment, University of Glasgow, Glasgow, UK
| | - Simon Kennedy
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Keith Oldroyd
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Sean McGinty
- Division of Biomedical Engineering, University of Glasgow, Glasgow, UK; Glasgow Computational Engineering Centre, Division of Infrastructure and Environment, University of Glasgow, Glasgow, UK.
| |
Collapse
|
3
|
Caminati G, Procacci P. Mounting evidence of FKBP12 implication in neurodegeneration. Neural Regen Res 2020; 15:2195-2202. [PMID: 32594030 PMCID: PMC7749462 DOI: 10.4103/1673-5374.284980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/18/2020] [Accepted: 03/24/2020] [Indexed: 12/25/2022] Open
Abstract
Intrinsically disordered proteins, such as tau or α-synuclein, have long been associated with a dysfunctional role in neurodegenerative diseases. In Alzheimer's and Parkinson's' diseases, these proteins, sharing a common chemical-physical pattern with alternating hydrophobic and hydrophilic domains rich in prolines, abnormally aggregate in tangles in the brain leading to progressive loss of neurons. In this review, we present an overview linking the studies on the implication of the peptidyl-prolyl isomerase domain of immunophilins, and notably FKBP12, to a variety of neurodegenerative diseases, focusing on the molecular origin of such a role. The involvement of FKBP12 dysregulation in the aberrant aggregation of disordered proteins pinpoints this protein as a possible therapeutic target and, at the same time, as a predictive biomarker for early diagnosis in neurodegeneration, calling for the development of reliable, fast and cost-effective detection methods in body fluids for community-based screening campaigns.
Collapse
Affiliation(s)
- Gabriella Caminati
- Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino, Italy
- Center for Colloid and Surface Science (CSGI), University of Florence, Sesto Fiorentino, Italy
| | - Piero Procacci
- Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
4
|
Schwartz SM, Virmani R, Majesky MW. An update on clonality: what smooth muscle cell type makes up the atherosclerotic plaque? F1000Res 2018; 7:F1000 Faculty Rev-1969. [PMID: 30613386 PMCID: PMC6305222 DOI: 10.12688/f1000research.15994.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
Almost 50 years ago, Earl Benditt and his son John described the clonality of the atherosclerotic plaque. This led Benditt to propose that the atherosclerotic lesion was a smooth muscle neoplasm, similar to the leiomyomata seen in the uterus of most women. Although the observation of clonality has been confirmed many times, interest in the idea that atherosclerosis might be a form of neoplasia waned because of the clinical success of treatments for hyperlipemia and because animal models have made great progress in understanding how lipid accumulates in the plaque and may lead to plaque rupture. Four advances have made it important to reconsider Benditt's observations. First, we now know that clonality is a property of normal tissue development. Second, this is even true in the vessel wall, where we now know that formation of clonal patches in that wall is part of the development of smooth muscle cells that make up the tunica media of arteries. Third, we know that the intima, the "soil" for development of the human atherosclerotic lesion, develops before the fatty lesions appear. Fourth, while the cells comprising this intima have been called "smooth muscle cells", we do not have a clear definition of cell type nor do we know if the initial accumulation is clonal. As a result, Benditt's hypothesis needs to be revisited in terms of changes in how we define smooth muscle cells and the quite distinct developmental origins of the cells that comprise the muscular coats of all arterial walls. Finally, since clonality of the lesions is real, the obvious questions are do these human tumors precede the development of atherosclerosis, how do the clones develop, what cell type gives rise to the clones, and in what ways do the clones provide the soil for development and natural history of atherosclerosis?
Collapse
Affiliation(s)
| | - Renu Virmani
- CV Path Institute, Gaithersberg, Maryland, 20878, USA
| | - Mark W. Majesky
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital Research Institute, Seattle, WA, 98112, USA
| |
Collapse
|
5
|
Tzafriri AR, Garcia-Polite F, Li X, Keating J, Balaguer JM, Zani B, Bailey L, Markham P, Kiorpes TC, Carlyle W, Edelman ER. Defining drug and target protein distributions after stent-based drug release: Durable versus deployable coatings. J Control Release 2018; 274:102-108. [PMID: 29421608 DOI: 10.1016/j.jconrel.2018.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 12/31/2017] [Accepted: 02/04/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Innovations in drug eluting stent designs make it increasingly important to develop models for differentiating performance through spatial definition of drug, receptor binding and cell state. METHODS Two designs of sirolimus analog eluting stents were implanted into porcine coronary arteries for 28, 60 or 90 days (n = 9/time point), durable coating (Xience) and deployable absorbable coating (MiStent). Explanted arteries were evaluated for drug content (n = 3/time point) by LC-MS/MS and for drug and target protein (mTOR) distributions by immunofluorescence (IF, n = 6/time point). A computational model was developed to predict drug release and arterial distribution maps. RESULTS Both stents released the majority of drug load by 28 days, with different tissue retention efficiencies (91.4 ± 4.9% MiStent versus 21.5 ± 1.9% Xience, P < 0.001). Computational modeling of MiStent coating deployment and microcrystal dissolution recapitulated in vivo drug release and net tissue content and predicted that >98.5% of deployed drug remains crystalline through 90 days. Immunofluorescence and computational modeling showed peristrut drug localization for both stents, with similar peaks, but high interstrut levels only at sites of coating deployment from the absorbable coating. Co-localization of mTOR-IF with drug-IF for both devices showed persistent drug effects, though with differential drug-receptor pharmacokinetics. CONCLUSIONS Immunofluorescence and computational modeling provide insights into drug distribution and binding status that can help differentiate drug delivery technologies. Herein we found that tissue deployment of slow dissolving crystalline drug particles results in temporally and spatially more uniform drug delivery to interstrut zones that might otherwise be under-dosed without excess peristrut drug.
Collapse
Affiliation(s)
| | | | - Xiaojian Li
- CBSET Inc., 500 Shire Way, Lexington, MA, USA
| | | | | | - Brett Zani
- CBSET Inc., 500 Shire Way, Lexington, MA, USA
| | - Lynn Bailey
- CBSET Inc., 500 Shire Way, Lexington, MA, USA
| | | | | | | | - Elazer R Edelman
- IMES, MIT, 77 Massachusetts Avenue, Cambridge, MA, USA; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Daniel JM, Dutzmann J, Brunsch H, Bauersachs J, Braun-Dullaeus R, Sedding DG. Systemic application of sirolimus prevents neointima formation not via a direct anti-proliferative effect but via its anti-inflammatory properties. Int J Cardiol 2017; 238:79-91. [DOI: 10.1016/j.ijcard.2017.03.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 01/06/2017] [Accepted: 03/12/2017] [Indexed: 01/15/2023]
|
7
|
Lekshmi KM, Che HL, Cho CS, Park IK. Drug- and Gene-eluting Stents for Preventing Coronary Restenosis. Chonnam Med J 2017; 53:14-27. [PMID: 28184335 PMCID: PMC5299126 DOI: 10.4068/cmj.2017.53.1.14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 12/19/2022] Open
Abstract
Coronary artery disease (CAD) has been reported to be a major cause of death worldwide. Current treatment methods include atherectomy, coronary angioplasty (as a percutaneous coronary intervention), and coronary artery bypass. Among them, the insertion of stents into the coronary artery is one of the commonly used methods for CAD, although the formation of in-stent restenosis (ISR) is a major drawback, demanding improvement in stent technology. Stents can be improved using the delivery of DNA, siRNA, and miRNA rather than anti-inflammatory/anti-thrombotic drugs. In particular, genes that could interfere with the development of plaque around infected regions are conjugated on the stent surface to inhibit neointimal formation. Despite their potential benefits, it is necessary to explore the various properties of gene-eluting stents. Furthermore, multifunctional electronic stents that can be used as a biosensor and deliver drug- or gene-based on physiological condition will be a very promising way to the successful treatment of ISR. In this review, we have discussed the molecular mechanism of restenosis, the use of drug- and gene-eluting stents, and the possible roles that these stents have in the prevention and treatment of coronary restenosis. Further, we have explained how multifunctional electronic stents could be used as a biosensor and deliver drugs based on physiological conditions.
Collapse
Affiliation(s)
- Kamali Manickavasagam Lekshmi
- Department of Biomedical Sciences and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea
| | - Hui-Lian Che
- Department of Biomedical Sciences and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea.; Heart Research Centre, Chonnam National University Hospital, Gwangju, Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
8
|
Arellano-Orden E, Serrano C, Montes-Worboys A, Sánchez-López V, Laborda A, Lostalé F, Lahuerta C, Rodríguez-Panadero F, de Gregorio MÁ. Stent-induced tracheal stenosis can be predicted by IL-8 expression in rabbits. Eur J Clin Invest 2017; 47:84-92. [PMID: 27873304 DOI: 10.1111/eci.12706] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/18/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Bare metal stents may cause complications like fibrous encapsulation, granulation and tracheal stenosis. We investigated the behaviour of three commercially available stents in vivo (rabbits) and in vitro (coculture of those stents with epithelial and fibroblast cell lines). Also, we investigated whether development of tracheal stenosis could be predicted by any biological marker. MATERIALS AND METHODS The tracheae of 30 rabbits were implanted with either nitinol stents, with or without paclitaxel elution, or a cobalt-based stent. An additional ten rabbits underwent mock implantation (controls). Serial peripheral venous blood samples were taken throughout the study, and several cytokines measured. Animals were euthanized on day 90, with immediate tracheal endoscopy and lavage performed, then necropsy. RESULTS Rabbits with cobalt-based stent exhibited more inflammation and the highest stenosis incidence, with reduced survival. Both in vivo and in vitro, this stent induced higher IL-8 levels than nitinol stents. Most important, the presence of stent-induced tracheal stenosis was closely associated to increase in IL-8 expression in blood just 1 day after tracheal stent implantation: a 1·19-fold increase vs. baseline had 83% sensitivity, 83% specificity, 77% positive predictive value, 88% negative predictive value and 83% accuracy to predict development of stenosis. CONCLUSIONS The cobalt-based stent had the highest incidence of tracheal inflammation and stenosis. On the other hand, the paclitaxel-eluting nitinol stent did not prevent those complications and provoked a marked reaction compared with the bare nitinol stent. Early increase in IL-8 expression in blood after stent implantation could predict development of tracheal stenosis in rabbits.
Collapse
Affiliation(s)
- Elena Arellano-Orden
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Carolina Serrano
- Minimally Invasive Techniques Research Group (GITMI), University of Zaragoza, Zaragoza, Spain
| | - Ana Montes-Worboys
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Verónica Sánchez-López
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Alicia Laborda
- Minimally Invasive Techniques Research Group (GITMI), University of Zaragoza, Zaragoza, Spain
| | - Fernando Lostalé
- Minimally Invasive Techniques Research Group (GITMI), University of Zaragoza, Zaragoza, Spain
| | - Celia Lahuerta
- Minimally Invasive Techniques Research Group (GITMI), University of Zaragoza, Zaragoza, Spain
| | - Francisco Rodríguez-Panadero
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Ángel de Gregorio
- Minimally Invasive Techniques Research Group (GITMI), University of Zaragoza, Zaragoza, Spain.,Interventional Radiology, Hospital Clinico Universitario, University of Zaragoza, Zaragoza, Spain.,CIBER Bioengineering, Biomaterials and Nanomedicine (BBB), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
9
|
Palmaz JC. Local Endovascular Delivery, Gene Therapy, and Cell Transplantation for Peripheral Arterial Disease. J Endovasc Ther 2016; 11 Suppl 2:II200-206. [PMID: 15760261 DOI: 10.1177/15266028040110s617] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Advances in catheter technology, gene identification, and cell biology may provide novel treatment options for patients with peripheral arterial disease (PAD) who are not candidates for standard revascularization procedures. Animal studies and recent results in human beings suggest that transfer of growth factors or regulatory genes and transplantation of progenitor cells may provide novel therapy options by inducing therapeutic angiogenesis or by inhibiting restenosis. This review will discuss the development of a variety of catheters for localized endovascular delivery, as well as the various cellular and genetic strategies that exist to restore blood flow to ischemic tissue and to reduce neointimal hyperplasia.
Collapse
Affiliation(s)
- Julio C Palmaz
- The University of Texas Health Science Center, San Antonio, Texas 78229, USA.
| |
Collapse
|
10
|
Levy E, Boulos A, Bendok B, Horowitz M, Kim S, Qureshi A, Guterman L, Hopkins L. Intracranial Stenting for Cerebrovascular Pathology. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/197140090301600118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
After reading this article, the participant should be able to: Describe the use of stent-assisted angioplasty for intracranial atherosclerotic disease. Recall the present status of stent-assisted managment of intracranial aneurysms. Describe the role of stenting and its technical aspects in the treatment of arteriovenous fistulae and acute stroke.
Collapse
Affiliation(s)
- E.I. Levy
- Dr. Levy is President, Department of Neurosurgery, University of Pittsburgh Medical Center-Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA 15213
| | - A.S. Boulos
- Dr. Boulos is Assistant Instructor of Clinical Neurosurgery
| | - B.R. Bendok
- Dr. Bendok is Assistant Professor of Neurosurgery
| | - M.B. Horowitz
- Dr. Horowitz is Associate Professor of Neurosurgery and Radiology, Departments of Neurological Surgery and Radiology, University of Pittsburgh Medical Center-Presbyterian University Hospital, 200 Lothrop Street, Pittsburgh PA 15213
| | - S.H. Kim
- Dr. Kim is Assistant Professor of Neurosurgery
| | - A.I. Qureshi
- Dr. Qureshi is Assistant Professor of Neurosurgery and Professor of Neurology
| | | | - L.N. Hopkins
- Dr. Hopkins is Director, Department of Neurosurgery and Toshiba Stroke Research Center, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Department of Neurosurgery, 3 Gates Circle, Buffalo NY 14209-1194
| |
Collapse
|
11
|
Tzafriri AR, Groothuis A, Price GS, Edelman ER. Stent elution rate determines drug deposition and receptor-mediated effects. J Control Release 2012; 161:918-26. [PMID: 22642931 DOI: 10.1016/j.jconrel.2012.05.039] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 05/16/2012] [Accepted: 05/19/2012] [Indexed: 10/28/2022]
Abstract
Drug eluting stent designs abound and yet the dependence of efficacy on drug dose and elution duration remains unclear. We examined these issues within a mathematical framework of arterial drug distribution and receptor binding following stent elution. Model predictions that tissue content linearly tracks stent elution rate were validated in porcine coronary artery sirolimus-eluting stents implants. Arterial content varied for stent types, progressively declining from its Day 1 peak and tracking with rate-limiting drug elution--near zero-order release was three-fold more efficient at depositing drug in the stented lesion than near first-order release. In vivo data were consistent with an overabundance of non-specific sirolimus-binding sites relative to the specific receptors and to the delivered dose. The implication is that the persistence time of receptor saturation and effect is more sensitive to duration of elution than to eluted amount. Consequently, the eluted amount should be sufficiently high to saturate receptors at the target lesion, but dose escalation alone is an inefficient strategy for prolonging the duration of sirolimus deposition. Moreover, receptor saturating drug doses are predicted to be most efficacious when eluted from stents in a constant zero order fashion as this maximizes the duration of elution and receptor saturation.
Collapse
|
12
|
Wang CH, Hsieh IC, Cherng WJ, Chen CC, Tung TH, Lee JF, Lin SJ, Wang PN. Differentiation profile of peripheral blood-derived vascular progenitor cell predicts intimal hyperplasia after coronary stenting. Heart Vessels 2012; 27:10-19. [PMID: 21331618 DOI: 10.1007/s00380-011-0118-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 01/14/2011] [Indexed: 10/18/2022]
Abstract
In-stent restenosis is largely due to intimal hyperplasia (IH). The number of vascular progenitor cells (VPCs) mobilized at the acute phase after stenting is associated with IH. This study sought to determine whether the differentiation profile of VPC predicts the development of IH. Peripheral blood was collected in 58 patients after bare-metal stenting to culture VPCs. Intravascular ultrasound was performed to estimate the area of IH 6 months after stenting. VPC differentiation was determined using flow cytometry. VE-cadherin (VE-Cad) and α-smooth muscle actin (α-SMA) were used to identify endothelial and smooth muscle cell lineages, respectively. After culturing, VPCs differentiated into four different phenotypes (α-SMA(-)VE-Cad(+), α-SMA(+)VE-cad(high), α-SMA(+)VE-cad(low), and α-SMA(+)VE-Cad(-)). IH was correlated with gender (P = 0.04), smoking status (P = 0.04), reference diameter (P = 0.03), minimal lumen diameter (P = 0.03), stent area (P < 0.0001), and parameters in the VPC differentiation profile (P < 0.05). Multivariate analysis controlling for stent area, smoking status, and gender revealed that IH was positively and independently associated with the number of differentiated α-SMA(+)VE-Cad (low/-) VPCs (P < 0.0001), and the ratio of α-SMA(+)VE-Cad (low/-) VPCs to α-SMA(-)VE-Cad(+) VPCs (P = 0.001). These parameters in the VPC differentiation profile independently predicted the IH and provided additive information to traditional risk factors. In conclusion, the profile of VPC differentiation predicts the severity of post-stent IH and may be a potential tool in the future for clinicians to identify patients at risk of post-stent restenosis.
Collapse
MESH Headings
- Aged
- Angioplasty, Balloon, Coronary/adverse effects
- Angioplasty, Balloon, Coronary/instrumentation
- Animals
- Biomarkers/metabolism
- Cell Differentiation
- Cell Proliferation
- Cells, Cultured
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/pathology
- Coronary Artery Disease/therapy
- Coronary Restenosis/diagnostic imaging
- Coronary Restenosis/etiology
- Coronary Restenosis/pathology
- Coronary Vessels/diagnostic imaging
- Coronary Vessels/pathology
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Female
- Humans
- Hyperplasia
- Linear Models
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Middle Aged
- Multivariate Analysis
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Risk Assessment
- Risk Factors
- Stem Cells/metabolism
- Stem Cells/pathology
- Stents
- Taiwan
- Time Factors
- Treatment Outcome
- Tunica Intima/diagnostic imaging
- Tunica Intima/pathology
- Ultrasonography, Interventional
- Vascular System Injuries/pathology
Collapse
Affiliation(s)
- Chao-Hung Wang
- Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, 222 Mai Chin Road, Keelung, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Ganesh SK, Joo J, Skelding K, Mehta L, Zheng G, O'Neill K, Billings EM, Helgadottir A, Andersen K, Thorgeirsson G, Gudnason T, Geller NL, Simari RD, Holmes DR, O'Neill WW, Nabel EG. Time course analysis of gene expression identifies multiple genes with differential expression in patients with in-stent restenosis. BMC Med Genomics 2011; 4:20. [PMID: 21356094 PMCID: PMC3053213 DOI: 10.1186/1755-8794-4-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 02/28/2011] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The vascular disease in-stent restenosis (ISR) is characterized by formation of neointima and adverse inward remodeling of the artery after injury by coronary stent implantation. We hypothesized that the analysis of gene expression in peripheral blood mononuclear cells (PBMCs) would demonstrate differences in transcript expression between individuals who develop ISR and those who do not. METHODS AND RESULTS We determined and investigated PBMC gene expression of 358 patients undergoing an index procedure to treat in de novo coronary artery lesions with bare metallic stents, using a novel time-varying intercept model to optimally assess the time course of gene expression across a time course of blood samples. Validation analyses were conducted in an independent sample of 97 patients with similar time-course blood sampling and gene expression data. We identified 47 probesets with differential expression, of which 36 were validated upon independent replication testing. The genes identified have varied functions, including some related to cellular growth and metabolism, such as the NAB2 and LAMP genes. CONCLUSIONS In a study of patients undergoing bare metallic stent implantation, we have identified and replicated differential gene expression in peripheral blood mononuclear cells, studied across a time series of blood samples. The genes identified suggest alterations in cellular growth and metabolism pathways, and these results provide the basis for further specific functional hypothesis generation and testing of the mechanisms of ISR.
Collapse
Affiliation(s)
- Santhi K Ganesh
- National Heart, Lung, and Blood Institute (NHLBI), Division of Intramural Research, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Dubois C, Wijns W. The AXXESS™ Self-Expanding Biolimus A9™ Eluting Stent System for coronary bifurcation lesions. EUROINTERVENTION 2010; 6 Suppl J:J130-4. [DOI: 10.4244/eijv6supja21] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
15
|
Pallero MA, Talbert Roden M, Chen YF, Anderson PG, Lemons J, Brott BC, Murphy-Ullrich JE. Stainless steel ions stimulate increased thrombospondin-1-dependent TGF-beta activation by vascular smooth muscle cells: implications for in-stent restenosis. J Vasc Res 2009; 47:309-22. [PMID: 20016205 DOI: 10.1159/000265565] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Accepted: 06/02/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Despite advances in stent design, in-stent restenosis (ISR) remains a significant clinical problem. All implant metals exhibit corrosion, which results in release of metal ions. Stainless steel (SS), a metal alloy widely used in stents, releases ions to the vessel wall and induces reactive oxygen species, inflammation and fibroproliferative responses. The molecular mechanisms are unknown. TGF-beta is known to be involved in the fibroproliferative responses of vascular smooth muscle cells (VSMCs) in restenosis, and TGF-beta antagonists attenuate ISR. We hypothesized that SS ions induce the latent TGF-beta activator, thrombospondin-1 (TSP1), through altered oxidative signaling to stimulate increased TGF-beta activation and VSMC phenotype change. METHODS VSMCs were treated with SS metal ion cocktails, and morphology, TSP1, extracellular matrix production, desmin and TGF-beta activity were assessed by immunoblotting. RESULTS SS ions stimulate the synthetic phenotype, increased TGF-beta activity, TSP1, increased extracellular matrix and downregulation of desmin in VSMCs. Furthermore, SS ions increase hydrogen peroxide and decrease cGMP-dependent protein kinase (PKG) signaling, a known repressor of TSP1 transcription. Catalase blocks SS ion attenuation of PKG signaling and increased TSP1 expression. CONCLUSIONS These data suggest that ions from stent alloy corrosion contribute to ISR through stimulation of TSP1-dependent TGF-beta activation.
Collapse
Affiliation(s)
- Manuel A Pallero
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Tzafriri AR, Vukmirovic N, Kolachalama VB, Astafieva I, Edelman ER. Lesion complexity determines arterial drug distribution after local drug delivery. J Control Release 2009; 142:332-8. [PMID: 19925836 DOI: 10.1016/j.jconrel.2009.11.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 11/11/2009] [Indexed: 01/23/2023]
Abstract
Though stents are deployed in diseased arteries drug distribution has only been quantified in intact, non-diseased vessels. We correlated steady-state arterial drug distribution with tissue ultrastructure and composition in abdominal aortae from atherosclerotic human autopsy specimens and rabbits with lesions induced by dietary manipulation and controlled injury. Paclitaxel, everolimus, and sirolimus depositions in the human aortae were maximal in the media and scaled inversely with lipid content. Net tissue paclitaxel and everolimus levels were indistinguishable in mildly injured rabbit arteries independent of diet. Yet, serial sectioning of cryopreserved arterial segments demonstrated a differential transmural deposition pattern that was amplified with disease and correlated with the expression of their intracellular targets, tubulin and FKBP-12. Tubulin distribution and paclitaxel binding increased with vascular injury and macrophage infiltration, and were reduced with lipid content. Sirolimus analogs and their specific binding target FKBP-12 were less sensitive to alterations of diet in mildly injured arteries, presumably reflecting a faster transient response of FKBP-12 to injury. The data demonstrate that disease-induced changes in the distribution of drug-binding proteins and interstitial lipid alter the distribution of these drugs, forcing one to consider how disease might affect the evaluation and efficacy of the local release of these and like compounds.
Collapse
Affiliation(s)
- Abraham R Tzafriri
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | | | | | | | |
Collapse
|
17
|
Common signatures for gene expression in postnatal patients with patent arterial ducts and stented arteries. Cardiol Young 2009; 19:352-9. [PMID: 19538825 DOI: 10.1017/s1047951109004260] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The detailed molecular processes associated with postnatal remodelling of blood vessels are presently not understood. To characterize the response of the patients undergoing stenting of the patent arterial duct, we harvested samples of vascular tissue during surgical repair. Histological analysis of explanted ducts confirmed the patency of the ducts immediately after birth. As expected, a previously unstented duct that was examined 7 months after birth had become closed and ligamentous. Whole genome expression profiling of these samples showed that a large fraction, over 10%, of the gene sequences examined were expressed differentially between the samples taken from patients with open as opposed to the ligamentous duct. Interestingly, in 2 patients in whom closure was prevented by insertion of stents, one showed an expression profile that was similar to that of the patient initially having an unstented open duct, whereas the other was more closely related to the profile of the patient with a duct that had become ligamentous. Moreover, in 2 specimens obtained from patients with stented pulmonary arteries, a large fraction of the genes that were differentially expressed were identical to the pattern seen in the samples from the patients with open ducts. The gene regulation appeared to be independent of the nature of the respective malformations, and the site of implantation of the stents. These findings suggest that a set of differentially expressed genes are indicative for a transcriptional programme in neonatal remodelling of the arterial duct, which may also take place in patients in whom ductal closure is prevented by stents, or in those with stented pulmonary arteries. The differentially expressed genes included a significant number of extracellular matrix synthetic genes, and could therefore be predictive for vascular remodelling and neointimal formation.
Collapse
|
18
|
Achrol AS, Guzman R, Lee M, Steinberg GK. Pathophysiology and genetic factors in moyamoya disease. Neurosurg Focus 2009; 26:E4. [PMID: 19335130 DOI: 10.3171/2009.1.focus08302] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Moyamoya disease is an uncommon cerebrovascular condition characterized by progressive stenosis of the bilateral internal carotid arteries with compensatory formation of an abnormal network of perforating blood vessels providing collateral circulation. The etiology and pathogenesis of moyamoya disease remain unclear. Evidence from histological studies, proteomics, and endothelial progenitor cell analyses suggests new theories underlying the cause of vascular anomalies, including moyamoya disease. Familial moyamoya disease has been noted in as many as 15% of patients, indicating an autosomal dominant inheritance pattern with incomplete penetrance. Genetic analyses in familial moyamoya disease and genome-wide association studies represent promising strategies for elucidating the pathophysiology of this condition. In this review, the authors discuss recent studies that have investigated possible mechanisms underlying the etiology of moyamoya disease, including stem cell involvement and genetic factors. They also discuss future research directions that promise not only to offer new insights into the origin of moyamoya disease but to enhance our understanding of new vessel formation in the CNS as it relates to stroke, vascular anomalies, and tumor growth.
Collapse
Affiliation(s)
- Achal S Achrol
- Departments of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, California 94305-5487, USA
| | | | | | | |
Collapse
|
19
|
Vascular progenitor cells and translational research: the role of endothelial and smooth muscle progenitor cells in endogenous arterial remodelling in the adult. Clin Sci (Lond) 2009; 116:283-99. [PMID: 19138170 DOI: 10.1042/cs20080001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There has been much recent research into the therapeutic use of stem and progenitor cells for various diseases. Alongside this, there has also been considerable interest in the normal roles that endogenous precursor cells may play in both physiological and pathological settings. In the present review, we focus on two types of progenitor cell which are of potential relevance to vascular homoeostasis, namely the EPC (endothelial progenitor cell) and the smooth muscle progenitor cell. We discuss evidence for their existence and sources in adults, and the various techniques currently used to identify these cells. We examine data obtained from studies using different methods of progenitor identification and relate these to each other, in order to provide a framework in which to interpret the literature in this area. We review evidence for the influence of these vascular progenitor cells upon vascular function and the development and progression of atherosclerosis.
Collapse
|
20
|
Zhao L, Ding T, Cyrus T, Cheng Y, Tian H, Ma M, Falotico R, Praticò D. Low-dose oral sirolimus reduces atherogenesis, vascular inflammation and modulates plaque composition in mice lacking the LDL receptor. Br J Pharmacol 2009; 156:774-85. [PMID: 19220291 DOI: 10.1111/j.1476-5381.2008.00080.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Chronic proliferative responses of different vascular cell types have been involved in the pathogenesis of atherosclerosis. However, their functional role remains to be established. Sirolimus reduces neointimal proliferation after balloon angioplasty and chronic graft vessel disease. These studies were undertaken to investigate the effects of this anti-proliferative drug on atherogenesis. EXPERIMENTAL APPROACH Low-density lipoprotein receptor-deficient (LDL r-KO) mice on a cholesterol-rich diet were randomized to receive placebo or sirolimus (0.1; 0.3; or 1 mg.kg(-1)) in their diet for 8 or 16 weeks. RESULTS In both studies, plasma levels of the drug increased in a dose-dependent fashion, animals gained weight normally and, among groups, plasma lipids levels did not differ significantly. Compared with placebo, plasma levels of interleukin-6, monocyte chemoattractant protein-1, interferon gamma, tumour necrosis factor alpha and CD40, and their mRNA levels in aortic tissue were significantly reduced in sirolimus-treated mice. This effect resulted in a significant and dose-dependent reduction in atherosclerotic lesions, in both the root and aortic tree. Also these lesions contained less monocyte/macrophages and smooth muscle cells, but more collagen. CONCLUSIONS AND IMPLICATIONS The present results demonstrated that at low doses, sirolimus was an effective and safe anti-atherogenic agent in the LDL r-KO mice. It attenuated the progression of atherosclerosis and modulated the plaque phenotype by reducing the pro-inflammatory vascular responses typical of the disease.
Collapse
Affiliation(s)
- L Zhao
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Klein CA, Zohlnhöfer D, Petat-Dutter K, Wendler N. Gene expression analysis of a single or few cells. ACTA ACUST UNITED AC 2008; Chapter 11:Unit 11.8. [PMID: 18428372 DOI: 10.1002/0471142905.hg1108s44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The need to analyze rare cells is based on the nature of tissue differentiation and regeneration, the initiation and propagation of disease processes in multicellular organisms, and the functional diversity of individual cells. Gene transcription is the most important regulatory mechanism by which a phenotype and functional state of a cell is determined. Therefore, procedures for the qualitative and quantitative assessment of mRNA abundance are important. This unit presents a protocol for semi-quantitative analysis of gene expression of a single cell and quantitative representation of expressed genes from >10 to 30 cells. A basic protocol for array hybridization on nylon filters is provided because such filters are available in every laboratory. Tissue samples contain many different cell types in variable amounts, so their analysis may require microdissection; a protocol for obtaining cryosections is given. Finally, a simple procedure to prepare the data for statistical analysis is also provided.
Collapse
|
22
|
Zohlnhöfer D, Nührenberg TG, Haas F, Bengel F, Schömig A, Baeuerle PA, Schwaiger M. Myocardial gene expression of matched hibernating and control tissue from patients with ischemic left ventricular dysfunction. Heart Vessels 2008; 23:230-42. [DOI: 10.1007/s00380-007-1035-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 11/30/2007] [Indexed: 12/26/2022]
|
23
|
Wang CH, Cherng WJ, Verma S. Drawbacks to stem cell therapy in cardiovascular diseases. Future Cardiol 2008; 4:399-408. [DOI: 10.2217/14796678.4.4.399] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Stem cells seem to have unlimited potential for repairing injured tissues derived from cardiovascular diseases. Much as the initial euphoria over preclinical models has ushered in some skepticism, several reports have advised caution against over exuberance, as cellular therapy has both theoretical and reported safety concerns. Embryonic stem cells, skeletal myoblasts, bone marrow-derived stem cells and mesenchymal stem cells are current candidates for cell therapy in end-stage cardiovascular diseases. However, before large-scale clinical trials can take place, a few safety concerns have to be clarified, such as atherogenesis, postangioplasty or stenting restenosis, tumorigenesis, stem cell metastasis, stem cell-mobilized cytokine-related complications and arrhythmogenesis. In this review, potential ways to overcome these issues are discussed, including medical and gene manipulations, dedicated cell-purification techniques, antiarrhythmic cell therapy design, and new concepts such as using natural constructs. With these safety issues under control, stem cell therapy still has a promising future in the next decade.
Collapse
Affiliation(s)
- Chao-Hung Wang
- Chang Gung University College of Medicine, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, 222 Mai-Chin Road, Keelung, Taiwan
| | - Wen-Jin Cherng
- Chang Gung University College of Medicine, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Subodh Verma
- St Michael’s Hospital, Division of Cardiovascular & Thoracic Surgery, Toronto, Canada
| |
Collapse
|
24
|
Klein CA, Zohlnhöfer D, Petat-Dutter K, Wendler N. Gene expression analysis of a single or few cells. ACTA ACUST UNITED AC 2008; Chapter 25:Unit 25B.8. [PMID: 18265324 DOI: 10.1002/0471142727.mb25b08s61] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The need to analyze rare cells is based on the nature of tissue differentiation and regeneration, the initiation and propagation of disease processes in multicellular organisms, and the functional diversity of individual cells. Gene transcription is the most important regulatory mechanism by which a phenotype and functional state of a cell is determined. Therefore, procedures for the qualitative and quantitative assessment of mRNA abundance are important. This unit presents a protocol for semi-quantitative analysis of gene expression of a single cell and quantitative representation of expressed genes from >10 to 30 cells. A basic protocol for array hybridization on nylon filters is provided because such filters are available in every laboratory. Tissue samples contain many different cell types in variable amounts, so their analysis may require microdissection; a protocol for obtaining cryosections is given. Finally, a simple procedure to prepare the data for statistical analysis is also provided.
Collapse
|
25
|
Diao Y, Guthrie S, Xia SL, Ouyang X, Zhang L, Xue J, Lee P, Grant M, Scott E, Segal MS. Long-term engraftment of bone marrow-derived cells in the intimal hyperplasia lesion of autologous vein grafts. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:839-48. [PMID: 18276778 DOI: 10.2353/ajpath.2008.070840] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Intimal hyperplasia of autologous vein grafts is a critical problem affecting the long-term patency of many types of vascular reconstruction. Within intimal hyperplasia lesions, smooth muscle cells are a major component, playing an essential role in the pathological process. Given that bone marrow-derived cells may differentiate into smooth muscle cells in the neointima of injured arteries, we hypothesized that the bone marrow may serve as a source for some of the smooth muscle cells within intimal hyperplasia lesions of vein grafts. To test this hypothesis, we used an established mouse model for intimal hyperplasia in wild-type mice that had been transplanted with bone marrow from a green fluorescent protein (GFP+/+) transgenic mouse. High-resolution confocal microscopy analysis performed 2 and 8 weeks after grafting demonstrated expression of GFP in 5.4 +/- 0.8% and 11.9 +/- 2.3%, respectively, of smooth muscle cells within intimal hyperplasia lesions. By 16 weeks, GFP expression in smooth muscle cells was not detected by immunohistochemistry; however, real-time PCR revealed that 20.2 +/- 1.7% of the smooth muscle cells captured from the neointima lesion by laser capture microdissection at 16 weeks contained GFP DNA. Our results suggest that bone marrow-derived cells differentiated into smooth muscle cells within the intimal lesion and may provide a novel clinical approach for decreasing intimal hyperplasia in vein grafts.
Collapse
Affiliation(s)
- Yanpeng Diao
- Division of Nephrology, Hypertension and Transplantation, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bauriedel G, Jabs A, Kraemer S, Nickenig G, Skowasch D. Neointimal expression of rapamycin receptor FK506-binding protein FKBP12: postinjury animal and human in-stent restenosis tissue characteristics. J Vasc Res 2007; 45:173-8. [PMID: 17962721 DOI: 10.1159/000110417] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 08/08/2007] [Indexed: 11/19/2022] Open
Abstract
Despite excellent clinical results for sirolimus (rapamycin)-eluting stents, the exact mechanisms of antirestenotic activity and affected cellular targets are incompletely understood. Therefore, we determined the presence and tem- porospatial expression pattern of FKBP12, the primary intracellular receptor of rapamycin, in rat carotid arteries after balloon injury, as well as in human in-stent restenosis and primary stable coronary atheroma. FKBP12 expression was assessed by immunohistochemistry. Rat carotid arteries revealed maximal expression in 57.7 +/- 4.0% of neointimal cells at day 7. A large proportion of these FKBP12+ cells showed luminally confined co-expression with dendritic cell markers. Despite a considerably thicker neointima at day 28, presence of FKBP12 decreased (8.5 +/- 1.9%, p = 0.02) with a scattered pattern in luminal and deep neointima. Likewise, human in-stent restenosis atherectomy specimens (time after stent implantation 2-12 months) revealed a comparable extent of cellular rapamycin receptor expression (9.3 +/- 1.0%) that significantly differed from that found in primary stable atheroma (1.3 +/- 0.4%, p < 0.001). In conclusion, the rapamycin receptor is predominantly present during early neointima formation, while mature neointimal atheromas show a relatively low expression without confinement to luminal areas. Co-expression of FKBP12 and dendritic cell markers suggests that dendritic cells may be another important target for early and long-term rapamycin effects.
Collapse
Affiliation(s)
- Gerhard Bauriedel
- Department of Internal Medicine II - Cardiology, University of Bonn, Bonn, Germany.
| | | | | | | | | |
Collapse
|
27
|
Charron T, Nili N, Strauss BH. The cell cycle: a critical therapeutic target to prevent vascular proliferative disease. Can J Cardiol 2007; 22 Suppl B:41B-55B. [PMID: 16498512 PMCID: PMC2780832 DOI: 10.1016/s0828-282x(06)70986-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Percutaneous coronary intervention is the preferred revascularization approach for most patients with coronary artery disease. However, this strategy is limited by renarrowing of the vessel by neointimal hyperplasia within the stent lumen (in-stent restenosis). Vascular smooth muscle cell proliferation is a major component in this healing process. This process is mediated by multiple cytokines and growth factors, which share a common pathway in inducing cell proliferation: the cell cycle. The cell cycle is highly regulated by numerous mechanisms ensuring orderly and coordinated cell division. The present review discusses current concepts related to regulation of the cell cycle and new therapeutic options that target aspects of the cell cycle.
Collapse
Affiliation(s)
| | | | - Bradley H Strauss
- Correspondence: Dr Bradley H Strauss, St Michael’s Hospital, 30 Bond Street, Toronto, Ontario M5B IW8. Telephone 416-864-5913, fax 416-864-5978, e-mail
| |
Collapse
|
28
|
Soda T, Suzuki H, Iso Y, Kusuyama T, Omori Y, Sato T, Shoji M, Koba S, Geshi E, Katagiri T. Bone marrow cells contribute to neointimal formation after stent implantation in swine. Int J Cardiol 2007; 121:44-52. [PMID: 17254651 DOI: 10.1016/j.ijcard.2006.10.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Accepted: 10/20/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND Recent studies suggest that neointimal cells in atherosclerotic lesions are partly derived from bone marrow (BM) cells. However, studies with large animal models have not yet clarified how BM cells contribute to neointimal formation in restenotic lesions. We examined the expression of CD34, a hematopoietic stem cell marker, in the neointima after coronary stent implantation in porcine. METHODS Pigs underwent balloon injury in the coronary arteries followed by stent implantation. The arteries were harvested at 3, 7, and 28 days after the stenting. The samples were used for immunohistochemistry for CD34, smooth muscle embryo (SMemb), alpha-smooth muscle cell actin (alpha-SMA), macrophage, c-kit, and AC133 antibodies. In morphometric analysis, each layer of vascular wall was calculated in the sections. RESULTS At 3 days, the expressions of CD34 and SMemb were minimal, and many macrophages were seen around the stent struts. At 7 days, co-expression of CD34 and SMemb was observed around the struts, and 11.5% of the neointimal cells were stained by CD34. In addition, c-kit positive cells and AC133 positive cells are detected in neointimal area. At 28 days, the neointima had thickened and expressed alpha-SMA rather than SMemb, and a few CD34-positive cells were detected. In morphometric analysis, luminal area/total vascular area was significantly smaller and intimal area/total vascular area was significantly bigger in 7 and 28 days than in the day of implantation. CONCLUSION BM cells of possibly hematopoietic origin partially contributed to neointimal formation after coronary stent implantation in a large animal model.
Collapse
Affiliation(s)
- Teruko Soda
- Third Department of Internal Medicine, Showa University School of Medicine, Hatanodai, Shinagawa-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Qian H, Yang Y, Li J, Huang J, Dou K, Yang G. The role of vascular stem cells in atherogenesis and post-angioplasty restenosis. Ageing Res Rev 2007; 6:109-127. [PMID: 17324640 DOI: 10.1016/j.arr.2007.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2006] [Revised: 09/16/2006] [Accepted: 01/19/2007] [Indexed: 12/20/2022]
Abstract
It is well known that atherosclerosis prevails in elderly populations as ageing acts as a recognized risk factor for this disease. Although the pathogenic factors leading to atherosclerosis are highly heterogeneous, traditionally speaking, the causative risk factors include hyperlipidemia, hypertension, diabetes mellitus and smoking, which can damage to endothelial function, and subsequently promote lipid penetration and inflammatory cell infiltration. Damaged endothelial cells (ECs) may be replaced by neighboring cell division, while damaged smooth muscle cells (SMCs) may be replaced by medial SMCs emigrating into the intima during atherogenesis. However, this standpoint is challenged by recent findings that vascular progenitor/stem cells (VPCs) may contribute to atherogenesis and post-angioplasty restenosis. VPCs are a group of primitive cells that have the potential to produce mature, functional cells in the vascular wall. VPCs residing in bone marrow, vascular wall or circulating in the peripheral blood may be stimulated by a variety of pathogenic factors. These stem cells then participate in regeneration, repair and remodeling of the injured arterial wall. This new concept may bring about a great breakthrough in understanding the pathogenesis of atherosclerosis and develop novel therapeutic strategies for coronary heart disease. This article will mainly review the role of VPCs in atherogenesis, thus providing a novel understanding about the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Haiyan Qian
- Department of Cardiology, Fuwai Hospital and Cardiovascular Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, 167 BeiLiShi Road, Beijing 100037, PR China
| | | | | | | | | | | |
Collapse
|
30
|
Wu-Wong JR, Nakane M, Ma J, Ruan X, Kroeger PE. VDR-mediated gene expression patterns in resting human coronary artery smooth muscle cells. J Cell Biochem 2007; 100:1395-405. [PMID: 17115413 DOI: 10.1002/jcb.21133] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vitamin D analogs such as paricalcitol and calcitriol that activate the vitamin D receptor (VDR) provide survival benefit for Stage 5 chronic kidney disease (CKD) patients, possibly associated with a decrease in cardiovascular (CV)-related incidents. Phenotypic changes of smooth muscle cells play an important role in CV disease. The role of vitamin D analogs in modulating gene expression in smooth muscle cells is still not well understood. In this study, DNA microarray analysis of approximately 22,000 different human genes was used to characterize the VDR-mediated gene expression profile in human coronary artery smooth muscle cells (CASMC) at rest. Cells in serum free medium were treated with 0.1 microM calcitriol (1alpha,25-dihydroxyvitamin D(3)) or paricalcitol (19-nor-1alpha,25-(OH)(2)D(2)) for 30 h. A total of 181 target genes were identified, with 103 genes upregulated and 78 downregulated (>two fold changes in either drug treatment group with P < 0.01). No significant difference was observed between calcitriol and paricalcitol. Target genes fell into various categories with the top five in cellular process, cell communication, signal transduction, development, and morphogenesis. Twenty-two selected genes linked to the CV system were also impacted. Real-time RT-PCR and/or Western blotting analysis were employed to confirm the expression patterns of selected genes such as 25-hydroxyvitamin D-24-hydroxylase, Wilms' tumor gene 1, transforming growth factorbeta3, plasminogen activator inhibitor-1, thrombospondin-1 (THBS1), and thrombomodulin (TM). This study provides insight into understanding the role of VDR in regulating gene expression in resting smooth muscle cells.
Collapse
MESH Headings
- Blotting, Western
- Bone Density Conservation Agents/pharmacology
- Calcitriol/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Proliferation/drug effects
- Cells, Cultured
- Cluster Analysis
- Coronary Vessels/cytology
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Dose-Response Relationship, Drug
- Electrophoresis, Polyacrylamide Gel
- Ergocalciferols/pharmacology
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Oligonucleotide Array Sequence Analysis
- Receptors, Calcitriol/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Vitamins/pharmacology
Collapse
Affiliation(s)
- J Ruth Wu-Wong
- Department of Integrative Pharmacology, Abbott Laboratories, Abbott Park, Illinois 60064, USA.
| | | | | | | | | |
Collapse
|
31
|
Li JM, Zhang X, Nelson PR, Odgren PR, Nelson JD, Vasiliu C, Park J, Morris M, Lian J, Cutler BS, Newburger PE. Temporal evolution of gene expression in rat carotid artery following balloon angioplasty. J Cell Biochem 2007; 101:399-410. [PMID: 17171642 DOI: 10.1002/jcb.21190] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The success of vascular intervention including angioplasty, stenting, and arterial bypass remains limited by negative remodeling resulted in lumen restenosis. This study was to characterize the global transcription profile reflecting concurrent events along arterial remodeling and neointima formation in a rat carotid artery balloon-injury model. Expression profiling of injured and control common carotid arteries on days 4, 7, 14 post-injury that mark the major pathohistological progression stages of neointimal formation were recorded on high-density oligonucleotide arrays. A subset of genes from microarray-based data was further studied using quantitative real time RT-PCR and in situ hybridization with sequential arterial samples from days 1 to 28 post-injury. The gene-encoded proteins were validated with Western blot. Besides temporal induction of a large cluster of genes over-represented by cell proliferation and macromolecule metabolism gene ontology categories, a fast-evolving inflammation could be demonstrated by the induction of Tgfb and other anti-inflammatory genes (e.g., C1qtnf3 (C1q and tumor necrosis factor related protein 3 (predicted))) and a shift from type 1 to 2 helper T cell response. The most significant signature of the induced neointimal profile is enrichment of genes functionally related to angiogenesis and extracellular matrix (ECM) remodeling (e.g., Spp1 (secreted phosphoprotein 1), CD44 (CD44 antigen), and Cxcl12 (chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1)). Some of the genes represent stress-responsive mesenchymal stromal cell cytokines. This study highlighted mesenchymal stromal cell cytokines-driven inflammatory extracellular matrix remodeling, as target processes for potential clinical therapeutic intervention.
Collapse
Affiliation(s)
- Jian-Ming Li
- Department of Surgery, Division of Vascular Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Blaes N, Elbaz M, Heitz F, Caussé E, Glock Y, Puel J, Bayard F. Differential display fingerprints: new approach to characterize smooth muscle cells and human coronary atherectomy tissues. ACTA ACUST UNITED AC 2007; 55:328-35. [PMID: 17611041 DOI: 10.1016/j.patbio.2007.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Accepted: 05/30/2007] [Indexed: 11/20/2022]
Abstract
AIM OF THE STUDY Smooth muscle cells build up the normal media and stabilize atherosclerotic lesions whereas an inflammatory component is determinant for unstable angina. Smooth muscle cells, currently identified by alpha-actin, present a phenotypic heterogeneity and alpha-actin can be reduced in pathology. We tried to characterize vascular cell types, particularly smooth muscle cells, and coronary atherosclerotic tissues, by random genes expression fingerprints. MATERIALS AND METHODS Expression fingerprints (cDNA electrophoresis) were performed by differential display reverse transcriptase-polymerase chain reaction. Variability of fingerprints was studied for a panel of arterial muscle cell phenotypes and comparisons were made with fingerprints from other cell types (endothelial cells and macrophages). The technique was then applied to human coronary atherectomy samples compared to control human arterial (mammary) smooth muscle. RESULTS Arterial smooth muscle cells fingerprints were overall similar whatever the cell phenotype (native contractile, dedifferentiated in culture or epithelioid). Moreover, with two primer pairs, the muscular fingerprints markedly differed from the endothelial and the monocytic fingerprints. Application of differential display to coronary atherectomy samples was feasible. Interestingly, the pathological tissues exhibited either smooth muscle-like or smooth muscle-divergent fingerprints. CONCLUSIONS Smooth muscle cells and inflammatory cells exhibited distinct differential display fingerprint patterns. Thus, a simple expression profile of arbitrary genes provides a molecular bar code tool (pattern signature) useful to characterize vascular cell cultures or tissues. The present work proposes a method to analyze coronary atherectomy samples which estimates their whole quality, muscular versus non muscular (inflammatory), this is of interest for clinical research.
Collapse
Affiliation(s)
- N Blaes
- Département cardiaque et rénal, institut de médecine moléculaire de Rangueil I2MR, U858, Inserm, 31432 Toulouse, France. blaes@toulouse
| | | | | | | | | | | | | |
Collapse
|
33
|
Heeneman S, Donners MMPC, Bai L, Daemen MJAP. Drug-induced immunomodulation to affect the development and progression of atherosclerosis: a new opportunity? Expert Rev Cardiovasc Ther 2007; 5:345-64. [PMID: 17338677 DOI: 10.1586/14779072.5.2.345] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Inflammation and cytokine pathways are crucial for the development and progression of atherosclerotic lesions. In this review, the hypothesis that immunomodulatory drugs provide a possible therapeutic modality for cardiovascular disease is evaluated. Therefore, after a short overview of the specific inflammatory pathways involved in atherosclerosis, literature on the effect of several immunomodulatory drugs, such as nonsteroidal anti-inflammatory drugs, specific cyclooxygenase inhibitors and immunosuppressive drugs, used currently in the prevention of rejection after organ transplant, on the development and progression of atherosclerosis is reviewed. In addition, the pleiotropic immunomodulatory effect of two established cardiovascular drugs (angiotensin-converting enzyme inhibitors and statins) is discussed.
Collapse
Affiliation(s)
- Sylvia Heeneman
- University of Maastricht, Department of Pathology, Cardiovascular Research Institute Maastricht, PO Box 5800, 6202 AZ Maastricht, The Netherlands.
| | | | | | | |
Collapse
|
34
|
Tölle M, Levkau B, Kleuser B, van der Giet M. Sphingosine-1-phosphate and FTY720 as anti-atherosclerotic lipid compounds. Eur J Clin Invest 2007; 37:171-9. [PMID: 17359484 DOI: 10.1111/j.1365-2362.2007.01776.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
All stages of atherosclerosis have been identified as a chronic vascular inflammatory disease. In the last few years there is increasing evidence that endogenous lysophospholipids such as sphingosine-1-phosphate (S1P) have potent anti-inflammatory properties. The S1P analogue FTY720 that has been developed as a potent, orally active, immunosuppressant in the field of transplantation and autoimmune disease has interesting effects on inflammatory processes in the arterial vessel wall. S1P targets five specific S1P receptors (S1P(1-5)), which are ubiquitously expressed. S1P(1-3) receptor expression is identified in arterial vessels. S1P and FTY720 show potent silencing effects on some vascular proinflammatory mechanisms in endothelial and vascular smooth muscle cells. In addition, the interaction of monocytes with the vessel wall is inhibited. As shown recently, FTY720 can effectively reduce the progression of atherosclerosis in apolipoprotein E-deficient mice having a high-cholesterol diet. It is not entirely clear which S1P receptor subtype is mainly involved in this process. However, it is currently speculated that the S1P(3) and probably the S1P(1) is involved in the anti-atherosclerotic effects of FTY720. This review summarizes the current knowledge about S1P- and FTY720-effects on mechanisms of vascular inflammatory disease. In addition S1P receptor subtypes are identified which might be interesting for molecular drug targeting.
Collapse
Affiliation(s)
- M Tölle
- Charite - Universitätsmedizin Berlin, Campus Benjamin Franklin, Med. Klinik mit Schwerpunkt Nephrologie, Berlin, Germany
| | | | | | | |
Collapse
|
35
|
Tanaka K, Sata M. Therapeutic Application of Bone Marrow-derived Progenitor Cells for Vascular Diseases: Magicbullets Having the Good Without the Bad? INT J GERONTOL 2007. [DOI: 10.1016/s1873-9598(08)70019-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
36
|
Rauma-Pinola T, Pääkkö P, Ilves M, Serpi R, Romppanen H, Vuolteenaho O, Ruskoaho H, Hautala T. Adrenomedullin gene transfer induces neointimal apoptosis and inhibits neointimal hyperplasia in injured rat artery. J Gene Med 2006; 8:452-8. [PMID: 16389603 DOI: 10.1002/jgm.865] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Arterial wall injury leads to inflammatory reaction and release of growth factors that may mediate intimal regrowth. It is hypothesized that the neointimal cells may originate from adventitial myofibroblasts, medial smooth muscle cells, or differentiated bone marrow derived cells. Adrenomedullin (AM), an auto/paracrine cardiovascular peptide that is secreted from fibroblasts, endothelial cells, and vascular smooth muscle cells, may have a regulatory role in the intimal regeneration. In order to investigate the role of AM in neointimal growth, stimulation of stem cell migration, and apoptosis, we overexpressed AM with recombinant adenovirus in a rat arterial injury model. The intimae were significantly thinner in the arteries treated with AM adenovirus compared to the control group. Intima/media ratios were 0.48 +/- 0.18 and 1.01 +/- 0.20 (P < 0.05) in the AM group and the control group, respectively. In addition, a significantly higher apoptotic index of neointimal cells was seen in the AM gene transfer group compared to the control (2.78 +/- 0.5 vs. 0.57 +/- 0.20, P < 0.01). The neointimal cells stained positive for alpha-smooth muscle actin and negative for desmin suggesting possible myofibroblast origin. Very few c-Kit+ or MDR1+ cells were detected 2 weeks after the injury. We conclude that AM overexpression inhibits neointimal growth. The inhibition is associated with enhanced apoptosis of the neointimal cells which may be of myofibroblast origin.
Collapse
Affiliation(s)
- Tanja Rauma-Pinola
- Department of Pharmacology and Toxicology, University of Oulu, Biocenter Oulu, Oulu, Finland
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Machan L. Clinical experience and applications of drug-eluting stents in the noncoronary vasculature, bile duct and esophagus. Adv Drug Deliv Rev 2006; 58:447-62. [PMID: 16713011 DOI: 10.1016/j.addr.2006.01.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2005] [Accepted: 01/31/2006] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To review the use of drug-eluting stents outside the coronary artery. FINDINGS The vast majority of research and clinical data on drug-eluting stents are from their use in coronary artery atherosclerosis; however, these devices can be used outside the coronary circulation in both vascular and nonvascular structures. In noncoronary arteries the principle indication for drug-eluting vascular stents is the same as in the coronary circulation, prevention of restenosis. Human experience has been essentially limited to trials or compassionate use; two small controlled studies and a number of small observational single center reports have been published, and there are trials in progress. To date the data have not been as compelling as in the coronary circulation. The physical characteristics of each vascular bed such as external compressive forces, blood flow rates, wall thickness relative to lumen size, and vessel wall composition differ significantly from the coronary circulation and each presents unique challenges to local drug delivery. Outside the vascular bed, the principle expected use is the prevention of tissue ingrowth after stent insertion in tubular structures such as the trachea, esophagus or bile ducts. CONCLUSIONS Considerable further study of drug-eluting stents will be required in each anatomic region to determine the ideal stent/drug combination and clinical appropriateness.
Collapse
Affiliation(s)
- Lindsay Machan
- Angiography and Interventional Radiology, Department of Radiology, Vancouver Hospital and Health Sciences Centre, Vancouver, BC, Canada.
| |
Collapse
|
38
|
Dichtl W, Stocker EM, Mistlberger K, Debbage P, Yan ZQ, Alber HF, Frick M, Dulak J, Pachinger O, Weidinger F. Countervailing effects of rapamycin (sirolimus) on nuclear factor-κB activities in neointimal and medial smooth muscle cells. Atherosclerosis 2006; 186:321-30. [PMID: 16185698 DOI: 10.1016/j.atherosclerosis.2005.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2004] [Revised: 08/04/2005] [Accepted: 08/12/2005] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Local application of rapamycin (sirolimus) by drug-eluting stents prevents lumen obliteration after angioplasty by inhibition of neointimal hyperplasia. The effects of rapamycin on neointimal smooth muscle cells (niSMC) which are responsible for the occurrence of restenosis have not been investigated so far. METHODS AND RESULTS Rat niSMC and medial SMC (mSMC) were obtained from balloon catheter-injured arteries. The niSMC exhibited higher basal NF-kappaB activity and TNF-alpha mRNA levels. Nuclear protein binding to NF-kappaB-DNA was attenuated in niSMC by incubation with rapamycin (0.1 and 1 microg/ml) for 24 and 48 h. In contrast in mSMC, 0.1 microg/ml rapamycin had no effect and at 1 microg/ml even increased nuclear protein binding to NF-kappaB-DNA. After 12 h incubation, rapamycin (0.001-10 microg/ml) induced IkappaB-alpha protein in niSMC, whereas in mSMC it stimulated IkappaB-alpha at much lower levels. Prolonged rapamycin treatment (1 microg/ml for 72 h) had no effect on TNF-alpha mRNA level and NF-kappaB activity in niSMC, whereas it led to their increase in mSMC. Vascular endothelial growth factor (VEGF) secretion was higher in mSMC than in niSMC; rapamycin decreased VEGF levels in both cell types. Ultrastructural analysis suggested that rapamycin caused early signs of degeneration in niSMC, but enhanced protein synthesis in mSMC. CONCLUSIONS This study shows that rapamycin influences the inflammatory phenotypes of SMC in opposite directions: it reduces the high basal NF-kappaB activity in niSMC and enhances NF-kappaB activity and TNF-alpha expression in mSMC. In addition, rapamycin inhibits VEGF production regardless of the phenotype of SMC. These findings shed light on molecular mechanisms and structural changes underlying therapeutic applications of rapamycin in prevention of restenosis, inhibition of chronic transplant arteriosclerosis and reduction of secondary malignoma formation due to immunosuppression.
Collapse
Affiliation(s)
- Wolfgang Dichtl
- Clinical Department of Cardiology, Medical University Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Buerke M, Guckenbiehl M, Schwertz H, Buerke U, Hilker M, Platsch H, Richert J, Bomm S, Zimmerman GA, Lindemann S, Mueller-Werdan U, Werdan K, Darius H, Weyrich AS. Intramural delivery of Sirolimus prevents vascular remodeling following balloon injury. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2006; 1774:5-15. [PMID: 16920414 PMCID: PMC2275912 DOI: 10.1016/j.bbapap.2006.04.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Revised: 04/04/2006] [Accepted: 04/24/2006] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Several studies have demonstrated that Sirolimus-eluting stents reduce restenosis in patients with coronary artery disease. Here, we tested whether direct delivery of Sirolimus into the vessel wall during balloon angioplasty can modify vascular remodeling over several weeks. METHODS AND RESULTS During angioplasty of the rabbit iliac artery we administered an intramural infusion of Sirolimus or its vehicle directly through a balloon catheter into the vessel wall. After 3 weeks neointimal formation was decreased (0.71+/-0.1 vs. 1.4+/-0.12 intima/media ratio), and this process was attributed to the inhibitory properties of Sirolimus on ECM deposition and smooth muscle cell proliferation. Sirolimus also significantly reduced the deposition of elastin, collagen III and fibronectin within the vascular wall. In parallel, proteomic profiles of arterial wall segments were obtained and 485 protein spots were consistently matched between non-dilated and dilated vessels. Differential expression of 12 proteins were observed between the groups and direct sequencing of digested peptides was performed. Local delivery of sirolimus during angioplasty attenuated the expression of structural proteins that included lamin A, vimentin, alpha-1-antitrypsin, and alpha-actin. CONCLUSIONS Local administration of Sirolimus during angioplasty prevents smooth muscle cell proliferation associated with vascular remodeling as well as the expression of extracellular matrix and structural proteins. Therefore, local injection of Sirolimus during balloon inflation may be an alternative therapeutic approach for preventing restenosis in small stenotic vessels (i.e., <2.5 mm).
Collapse
Affiliation(s)
- Michael Buerke
- Department of Medicine III, Martin-Luther-University, Ernst-Grube-Str. 40, 06097 Halle, and Department of Cardiology, Neu-Kölln-Hospital, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
McGillicuddy FC, O'Toole D, Hickey JA, Gallagher WM, Dawson KA, Keenan AK. TGF-beta1-induced thrombospondin-1 expression through the p38 MAPK pathway is abolished by fluvastatin in human coronary artery smooth muscle cells. Vascul Pharmacol 2006; 44:469-75. [PMID: 16624629 DOI: 10.1016/j.vph.2006.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 02/13/2006] [Accepted: 03/09/2006] [Indexed: 10/24/2022]
Abstract
Thrombospondin-1 (TSP-1) and transforming growth factor-beta1 (TGF-beta1) are both implicated in the pathogenesis of in-stent restenosis. This study evaluated the hypothesis that the HMG-CoA reductase inhibitor fluvastatin inhibits TGF-beta1 induced TSP-1 expression via inhibition of p38 mitogen activated protein kinase (MAPK) phosphorylation in human coronary artery smooth muscle cells (HCASMC) and may therefore have anti-restenosis potential. Fluvastatin significantly reduced TSP-1 mRNA and protein expression in HCASMC in a concentration-dependent manner with a significant reduction in expression observed after treatment with 0.25 microM fluvastatin. TGF-beta1 (5 ng/ml) induced phosphorylation of p38 MAPK and induced TSP-1 mRNA and protein expression in HCASMC. Fluvastatin abolished TGF-beta1-induced phosphorylation of p38 MAPK and TGF-beta1-induced TSP-1 expression. Blockade of the p38 MAPK pathway with the upstream inhibitor SB-203580 also abolished TGF-beta1-induced TSP-1 expression. We conclude that fluvastatin decreases expression of TSP-1 and abolishes the ability of TGF-beta1 to induce TSP-1 expression in HCASMC; this may be achieved by preventing signalling through the p38 MAPK pathway. Targeted delivery of fluvastatin may therefore be a useful therapeutic objective for prevention of the intimal hyperplasia associated with in-stent restenosis.
Collapse
MESH Headings
- Adult
- Cells, Cultured
- Coronary Restenosis/prevention & control
- Coronary Vessels/drug effects
- Coronary Vessels/enzymology
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Fatty Acids, Monounsaturated/pharmacology
- Fatty Acids, Monounsaturated/therapeutic use
- Fluvastatin
- Gene Expression Regulation
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use
- Imidazoles/pharmacology
- Indoles/pharmacology
- Indoles/therapeutic use
- MAP Kinase Signaling System
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Phosphorylation
- Pyridines/pharmacology
- RNA, Messenger/metabolism
- Thrombospondin 1/genetics
- Thrombospondin 1/metabolism
- Transforming Growth Factor beta/pharmacology
- Transforming Growth Factor beta1
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Fiona C McGillicuddy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
41
|
Gardell SE, Dubin AE, Chun J. Emerging medicinal roles for lysophospholipid signaling. Trends Mol Med 2006; 12:65-75. [PMID: 16406843 DOI: 10.1016/j.molmed.2005.12.001] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Revised: 11/21/2005] [Accepted: 12/09/2005] [Indexed: 02/06/2023]
Abstract
The two lysophospholipids (LPs) lysophosphatidic acid and sphingosine 1-phosphate (S1P) regulate diverse biological processes. Over the past decade, it has become clear that medically relevant LP activities are mediated by specific G protein-coupled receptors, implicating them in the etiology of a growing number of disorders. A new class of LP agonists shows promise for drug therapy: the experimental drug FTY720 is phosphorylated in vivo to produce a potent S1P receptor agonist (FTY720-P) and is currently in Phase III clinical trials for kidney transplantation and Phase II for multiple sclerosis. Recent genetic and pharmacological studies on LP signaling in animal disease models have identified new areas in which interventions in LP signaling might provide novel therapeutic approaches for the treatment of human diseases.
Collapse
Affiliation(s)
- Shannon E Gardell
- Department of Molecular Biology, Helen L. Dorris Child and Adolescent Neuropsychiatric Disorder Institute, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
42
|
Cilingiroglu M, Elliott J, Patel D, Tio F, Matthews H, McCasland M, Trauthen B, Elicker J, Bailey SR. Long-term effects of novel biolimus eluting DEVAX AXXESS plus nitinol self-expanding stent in a porcine coronary model. Catheter Cardiovasc Interv 2006; 68:271-9. [PMID: 16810698 DOI: 10.1002/ccd.20848] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The purpose of this study was to evaluate the long-term effects of the DEVAX AXXESS biolimus eluting stent (BES) in a porcine coronary model, compared with those of bare metal stent (BMS) and polymer only stent (POS) controls. BACKGROUND Excessive neointimal growth has been identified as a major cause of late failure of percutaneous coronary interventions. The effect of drug eluting from self-expanding stents for prevention of neointimal hyperplasia has not been studied before. The DEVAX AXXESS is a self-expanding nickel titanium stent, coated with antiproliferative compound-biolimus. METHODS Twenty juvenile farm swine, 25-35 kg in weight, 3-6 months in age were used. Each animal received a stent to the left anterior descending artery, left circumflex or right coronary arteries as permitted per anatomy. The chronic vascular response after BES implantation was compared with that after BMS and POS implantation at 28, 90, and 180 days follow-up. RESULTS The 28-day outcome by quantitative coronary angiography (QCA) showed significant increase in minimal luminal diameter (MLD) in the BES (MLD: 2.90 +/- 0.97, 2.39 +/- 0.90, 1.59 +/- 0.91; P = 0.009) compared with BMS and POS, respectively. By histomorphometric analysis, there was also a corresponding significant reduction in neointimal tissue proliferation in the BES (average neointimal area: 2.78 +/- 0.07, 5.46 +/- 0.66, 8.42 +/- 0.85; P = 0.002) compared with that in BMS and POS controls, respectively at 28-days follow-up. At 90 and 180 days, the mean neointimal area was not significantly different between the BES and the controls. CONCLUSIONS BES favorably modulates the neointimal tissue formation for 28 days, in the porcine coronary model. Long-term inhibition of neointimal hyperplasia is not sustained most likely because of the delayed cellular proliferation and inflammation in the vessel wall.
Collapse
Affiliation(s)
- Mehmet Cilingiroglu
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center in San Antonio, Texas 78229-3900, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sata M, Fukuda D, Tanaka K, Kaneda Y, Yashiro H, Shirakawa I. The role of circulating precursors in vascular repair and lesion formation. J Cell Mol Med 2005; 9:557-68. [PMID: 16202205 PMCID: PMC6741295 DOI: 10.1111/j.1582-4934.2005.tb00488.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The accumulation of smooth muscle cells (SMCs) plays a principal role in atherogenesis, post-angioplasty restenosis and transplantation-associated vasculopathy. Therefore, much effort has been expended in targeting the migration and proliferation of medial smooth muscle cells to prevent occlusive vascular remodeling. Recent evidence suggests that bone marrow-derived circulating precursors can also give rise to endothelial cells and smooth muscle cells that contribute to vascular repair, remodeling, and lesion formation under physiological and pathological conditions. This article overviews recent findings on circulating vascular progenitor cells and describes potential therapeutic strategies that target these cells to treat occlusive vascular diseases.
Collapse
Affiliation(s)
- Masataka Sata
- Department of Cardiovascular Medicine, University of Tokyo, Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | | | | | | | | | | |
Collapse
|
44
|
Cagiannos C, Abul-Khoudoud OR, DeRijk W, Shell DH, Jennings LK, Tolley EA, Handorf CR, Fabian TC. Rapamycin-coated expanded polytetrafluoroethylene bypass grafts exhibit decreased anastomotic neointimal hyperplasia in a porcine model. J Vasc Surg 2005; 42:980-8. [PMID: 16275457 DOI: 10.1016/j.jvs.2005.06.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Accepted: 06/20/2005] [Indexed: 11/29/2022]
Abstract
OBJECTIVE We tested the hypothesis that rapamycin coated onto, and eluted from, expanded polytetrafluoroethylene (ePTFE) grafts would diminish neointimal hyperplasia in a porcine model. METHODS Rapamycin (also called sirolimus) was coated onto the luminal surface of 6-mm-internal-diameter thin-walled ePTFE grafts by using an adhesive polymer that allows timed release of the drug. An adhesive polymer that allows timed release of rapamycin from ePTFE was developed with commercially available chemicals and applied on 6-mm ePTFE grafts. Graft integrity was characterized by scanning electron microscopy, and rapamycin levels were quantified by using high-performance liquid chromatography. Twenty-two mongrel pigs were randomized into three groups: untreated ePTFE (n = 6), adhesive-only coated ePTFE (n = 6), or adhesive- and rapamycin-coated ePTFE (n = 10). End-to-side unilateral aortoiliac bypasses were performed by using 6-mm-internal-diameter ePTFE grafts and standardized anastomotic lengths. Unilateral end-to-side aortoiliac ePTFE grafts (6-mm internal diameter) were inserted by using polypropylene sutures, 6-0 proximally and 7-0 distally; all anastomoses were 12 mm long. All animals received aspirin (325 mg orally) daily. All animals were given oral aspirin (325 mg) daily beginning on the day before surgery. At 28 days, the animals were killed, and the grafts were explanted in continuity with the adjacent aortic cuff and the outflow iliac artery. Variables compared between groups included graft patency, distal anastomotic length and cross-sectional narrowing, and intimal thickness at the arterial-graft junction indexed to the adjacent graft thickness. Microscopic analysis was performed with hematoxylin and eosin and Masson trichrome stains on paraffin sections. A pathologist blinded to experimental groups graded sections for collagen deposition, neointima formation, inflammatory cellular infiltrates, medial necrosis, and aneurysmal degeneration. RESULTS All animals survived until they were killed without clinical evidence of limb ischemia or graft infection. Preplanned t tests in the context of one-way analysis of variance showed no difference in outcome measures between the untreated ePTFE and adhesive-only coated ePTFE groups; therefore, they were combined in further comparisons with the adhesive- and rapamycin-coated ePTFE group. The Rapamycine eluting expanded polytetrafluoroethylene group had longer anastomoses (85.6% vs 60.6% of the initial anastomotic length maintained; P < .0001) and less cross-sectional narrowing in the outflow graft (16.2% vs 28.5%; P = .0007) when compared with the other two groups by using two-tailed Student t tests. There was no evidence of medial necrosis or aneurysmal degeneration. All patent grafts had complete endothelialization on hematoxylin and eosin sections. Rapamycin was detectable and quantifiable in the arterial wall at 28 days after implantation. CONCLUSIONS Rapamycin can be coated onto and eluted from ePTFE by using a nonionic polymer and a simple coating technique. At 4 weeks after implantation, the rapamycin-eluting ePTFE grafts demonstrate gross, pathologic, and morphometric features of diminished neointimal hyperplasia when compared with non-drug-eluting ePTFE. Four weeks after implantation in a porcine model, rapamycin-eluting ePTFE grafts demonstrated gross, pathologic, and morphometric features of diminished neointimal hyperplasia when compared with untreated and adhesive-only coated ePTFE grafts. CLINICAL RELEVANCE Rapamycin-eluting ePTFE grafts decrease neointimal hyperplasia in a porcine model. Further studies are needed to evaluate whether patency will be improved. Rapamycin-eluting ePTFE grafts may allow the use of prosthetic grafts in situations in which autologous vein is unavailable and in which neointimal hyperplasia is pronounced, such as in small-diameter (<6-mm) vessels typical of infrapopliteal interventions.
Collapse
Affiliation(s)
- Catherine Cagiannos
- Department of Surgery, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Scheller B, Grandt A, Wnendt S, Lorenz G, Böhm M, Nickenig G. Comparative study of tacrolimus and paclitaxel stent coating in the porcine coronary model. ACTA ACUST UNITED AC 2005; 94:445-52. [PMID: 15997345 DOI: 10.1007/s00392-005-0237-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Accepted: 01/25/2005] [Indexed: 11/28/2022]
Abstract
BACKGROUND Tacrolimus is a potent antiproliferative and immunosuppressive agent allowing for improved endothelial regeneration. The aim of our study was the preclinical evaluation of tacrolimus in a drug eluting nonerodable polymer stent system and its comparison with paclitaxel. METHODS AND RESULTS A total of 40 domestic pigs and 10 mini-pigs underwent coronary stenting with a follow-up time between 6 hours and 3 months. Stents were implanted in coronary arteries with an overstretch ratio of 1.2. After 3 days, a 1.73 microg/mm(2) coating produced tacrolimus tissue levels of 20 mumol/l in the coronary artery wall. Effective tissue concentrations were sustained for 28 days. Based on histomorphometric analysis, tacrolimus stent treated vessels had a reduced extent of neointima formation compared with controls at 28 days (-51% compared to control) but not at 3 months. High dose paclitaxel stent coating (1.44 microg/mm(2)) was complicated by unexpected deaths of pigs and thrombotic stent occlusion at control angiography. Long-term porcine data showed no persistent inhibition of neointimal growth by paclitaxel and tacrolimus stent coating. CONCLUSIONS Similar to paclitaxel, tacrolimus stent coating reduces neointimal proliferation in the porcine coronary model. However, dosing and long-term efficacy remains a critical issue in stent-based local drug delivery.
Collapse
Affiliation(s)
- B Scheller
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, 66421 Homburg/Saar, Germany.
| | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Alkawi A, Kirmani JF, Janjua N, Khatri I, Ahmed S, Divani AA, Qureshi AI. Advances in thrombolytics and mechanical devices for treatment of acute ischemic stroke. Neurol Res 2005; 27 Suppl 1:S42-9. [PMID: 16197823 DOI: 10.1179/016164105x25306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Stroke is a leading cause of death and disability. Recently, there have been advances in the treatment of acute ischemic stroke aimed at re-establishing blood flow to the affected area in an effort to save the ischemic penumbra surrounding the area of infarction. This is achieved by the use of thrombolytics intravenously or intra-arterially. The use of mechanical devices facilitates the function of pharmacological agents used in addition to minimizing the associated risks. In this review, we first discuss the therapeutic potentials and strategies employed in using different thrombolytics in management of acute ischemic stroke. Subsequently, we discuss the recent advances and therapeutic applications of mechanical devices in this field.
Collapse
Affiliation(s)
- Ammar Alkawi
- Zeenat Qureshi Stroke Research Center, Department of Neurology and Neurosciences, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Shih CM, Lin SJ, Su YY, Shih CC. Amorphous oxide--a platform for drug delivery. J Control Release 2005; 102:539-49. [PMID: 15681077 DOI: 10.1016/j.jconrel.2004.10.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2004] [Accepted: 10/15/2004] [Indexed: 11/18/2022]
Abstract
Usually, a drug is loaded onto the metallic surface of a medical device by applying a polymer layer containing the drug. Unfortunately, polymer coatings on the metallic surface may exhibit numerous problems after implantation, such as late thrombosis, inflammation, and restenosis. Current research was conducted to investigate whether a suitable oxide layer can be used as a polymer-free platform for drug loading, especially for cardiovascular stents. The loading of heparin onto, as well as eluting of heparin from, the amorphous oxide film on the 316LVM stainless steel wire was confirmed by experimental studies using cyclic voltammetry (CV), electrochemical impedance spectroscopy (EIS), electron spectroscopy for chemical analysis (ESCA), high-performance liquid chromatography (HPLC), and activated clotting time (ACT). Evidence shows that amorphous oxide can be an ideal substitute for the polymer coating of drug-loaded stents to minimize metallic corrosion, inflammation, late-thrombosis, and restenosis.
Collapse
Affiliation(s)
- Chun-Ming Shih
- Graduate Institute of Medical Sciences, School of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | | | | | | |
Collapse
|
49
|
Donners MMPC, Bot I, De Windt LJ, van Berkel TJC, Daemen MJAP, Biessen EAL, Heeneman S. Low-dose FK506 blocks collar-induced atherosclerotic plaque development and stabilizes plaques in ApoE-/- mice. Am J Transplant 2005; 5:1204-15. [PMID: 15888024 DOI: 10.1111/j.1600-6143.2005.00821.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Since atherosclerosis is a chronic inflammatory disease, we tested the hypothesis that the immunosuppressive drug FK506 would attenuate the development of atherosclerosis using a mouse model of collar-induced atherosclerosis. ApoE-/- mice were treated for 4 weeks with the immunosuppressive drug FK506 (0.05 mg/kg/day), yielding sustained blood levels (approximately 0.2 ng/mL) without systemic side effects. Atherosclerotic plaque development of FK506-treated mice was significantly reduced (63%) while plaque cell density was increased (52%) compared to controls. Importantly, FK506 also blocked progression of pre-existing atherosclerotic plaques. Plaque area of pre-existing plaques was 35% reduced by FK506. Cell density (35%) and collagen content (51%) were significantly increased, whereas necrotic core content was decreased (42%), indicating a more stable plaque morphology. Similar results were found during spontaneous atherosclerotic plaque development in ApoE-/- mice (treatment 17-25 weeks of age). Flow-cytometric analysis showed no peripheral effects on blood cell count or T-cell activation after FK506-treatment. In vitro, FK506 decreased vascular smooth muscle cell (VSMC) apoptosis and inhibited nuclear factor of activated T cells (NFAT)-luciferase reporter activity at concentrations in the range of the in vivo concentration. Low-dose FK506 inhibits collar-induced atherosclerotic plaque development and progression and induces more stable plaque phenotypes in ApoE-/- mice without any peripheral side effects.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/genetics
- Apolipoproteins E/physiology
- Apoptosis/drug effects
- Arteriosclerosis/pathology
- Arteriosclerosis/prevention & control
- Blood Cell Count
- Disease Models, Animal
- Immunosuppressive Agents/administration & dosage
- Luciferases/metabolism
- Lymphocyte Activation
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- NF-kappa B/metabolism
- Plaque, Amyloid/pathology
- Tacrolimus/administration & dosage
Collapse
Affiliation(s)
- Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
50
|
Opie SR, Dib N. Local endovascular delivery, gene therapy, and cell transplantation for peripheral arterial disease. J Endovasc Ther 2005. [PMID: 15760251 DOI: 10.1583/04-1319.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Advances in catheter technology, gene identification, and cell biology may provide novel treatment options for patients with peripheral arterial disease (PAD) who are not candidates for standard revascularization procedures. Animal studies and recent results in human beings suggest that transfer of growth factors or regulatory genes and transplantation of progenitor cells may provide novel therapy options by inducing therapeutic angiogenesis or by inhibiting restenosis. This review will discuss the development of a variety of catheters for localized endovascular delivery, as well as the various cellular and genetic strategies that exist to restore blood flow to ischemic tissue and to reduce neointimal hyperplasia.
Collapse
Affiliation(s)
- Shaun R Opie
- Department of Cell and Gene Research, Arizona Heart Institute and Foundation, Phoenix, Arizona 85016, USA.
| | | |
Collapse
|