1
|
De Ornelas B, Sucato V, Vadalà G, Buono A, Galassi AR. Myocardial Bridge and Atherosclerosis, an Intimal Relationship. Curr Atheroscler Rep 2024; 26:353-366. [PMID: 38822987 DOI: 10.1007/s11883-024-01219-1] [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] [Accepted: 05/18/2024] [Indexed: 06/03/2024]
Abstract
PURPOSE OF REVIEW This review investigates the relationship between myocardial bridges (MBs), intimal thickening in coronary arteries, and Atherosclerotic cardiovascular disease. It focuses on the role of mechanical forces, such as circumferential strain, in arterial wall remodeling and aims to clarify how MBs affect coronary artery pathology. REVIEW FINDINGS MBs have been identified as influential in modulating coronary artery intimal thickness, demonstrating a protective effect against thickening within the MB segment and an increase in thickness proximal to the MB. This is attributed to changes in mechanical stress and hemodynamics. Research involving arterial hypertension models and vein graft disease has underscored the importance of circumferential strain in vascular remodeling and intimal hyperplasia. Understanding the complex dynamics between MBs, mechanical strain, and vascular remodeling is crucial for advancing our knowledge of coronary artery disease mechanisms. This could lead to improved management strategies for cardiovascular diseases, highlighting the need for further research into MB-related vascular changes.
Collapse
Affiliation(s)
- Benjamin De Ornelas
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Vincenzo Sucato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giuseppe Vadalà
- Division of Cardiology, University Hospital "P. Giaccone", Palermo, Italy
| | - Andrea Buono
- Interventional Cardiology Unit, Cardiovascular Department, Fondazione Poliambulanza Institute, Brescia, Italy
| | - Alfredo Ruggero Galassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
2
|
Lemay SE, Montesinos MS, Grobs Y, Yokokawa T, Shimauchi T, Romanet C, Sauvaget M, Breuils-Bonnet S, Bourgeois A, Théberge C, Pelletier A, El Kabbout R, Martineau S, Yamamoto K, Ray AS, Lippa B, Goodwin B, Lin FY, Wang H, Dowling JE, Lu M, Qiao Q, McTeague TA, Moy TI, Potus F, Provencher S, Boucherat O, Bonnet S. Exploring Integrin α5β1 as a Potential Therapeutic Target for Pulmonary Arterial Hypertension: Insights from Comprehensive Multicenter Preclinical Studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.596052. [PMID: 38854025 PMCID: PMC11160677 DOI: 10.1101/2024.05.27.596052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by obliterative vascular remodeling of the small pulmonary arteries (PA) and progressive increase in pulmonary vascular resistance (PVR) leading to right ventricular (RV) failure. Although several drugs are approved for the treatment of PAH, mortality remains high. Accumulating evidence supports a pathological function of integrins in vessel remodeling, which are gaining renewed interest as drug targets. However, their role in PAH remains largely unexplored. We found that the arginine-glycine-aspartate (RGD)-binding integrin α5β1 is upregulated in PA endothelial cells (PAEC) and PA smooth muscle cells (PASMC) from PAH patients and remodeled PAs from animal models. Blockade of the integrin α5β1 or depletion of the α5 subunit resulted in mitotic defects and inhibition of the pro-proliferative and apoptosis-resistant phenotype of PAH cells. Using a novel small molecule integrin inhibitor and neutralizing antibodies, we demonstrated that α5β1 integrin blockade attenuates pulmonary vascular remodeling and improves hemodynamics and RV function in multiple preclinical models. Our results provide converging evidence to consider α5β1 integrin inhibition as a promising therapy for pulmonary hypertension. One sentence summary The α5β1 integrin plays a crucial role in pulmonary vascular remodeling.
Collapse
|
3
|
Rama E, Mohapatra SR, Melcher C, Nolte T, Dadfar SM, Brueck R, Pathak V, Rix A, Gries T, Schulz V, Lammers T, Apel C, Jockenhoevel S, Kiessling F. Monitoring the Remodeling of Biohybrid Tissue-Engineered Vascular Grafts by Multimodal Molecular Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105783. [PMID: 35119216 PMCID: PMC8981893 DOI: 10.1002/advs.202105783] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Indexed: 06/10/2023]
Abstract
Tissue-engineered vascular grafts (TEVGs) with the ability to grow and remodel open new perspectives for cardiovascular surgery. Equipping TEVGs with synthetic polymers and biological components provides a good compromise between high structural stability and biological adaptability. However, imaging approaches to control grafts' structural integrity, physiological function, and remodeling during the entire transition between late in vitro maturation and early in vivo engraftment are mandatory for clinical implementation. Thus, a comprehensive molecular imaging concept using magnetic resonance imaging (MRI) and ultrasound (US) to monitor textile scaffold resorption, extracellular matrix (ECM) remodeling, and endothelial integrity in TEVGs is presented here. Superparamagnetic iron-oxide nanoparticles (SPION) incorporated in biodegradable poly(lactic-co-glycolic acid) (PLGA) fibers of the TEVGs allow to quantitatively monitor scaffold resorption via MRI both in vitro and in vivo. Additionally, ECM formation can be depicted by molecular MRI using elastin- and collagen-targeted probes. Finally, molecular US of αv β3 integrins confirms the absence of endothelial dysfunction; the latter is provocable by TNF-α. In conclusion, the successful employment of noninvasive molecular imaging to longitudinally evaluate TEVGs remodeling is demonstrated. This approach may foster its translation from in vitro quality control assessment to in vivo applications to ensure proper prostheses engraftment.
Collapse
Affiliation(s)
- Elena Rama
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Saurav Ranjan Mohapatra
- Department of Biohybrid & Medical TextilesInstitute of Applied Medical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Christoph Melcher
- Institute for Textile Technology RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Teresa Nolte
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Seyed Mohammadali Dadfar
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Ramona Brueck
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Vertika Pathak
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Anne Rix
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Thomas Gries
- Institute for Textile Technology RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Volkmar Schulz
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Christian Apel
- Department of Biohybrid & Medical TextilesInstitute of Applied Medical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical TextilesInstitute of Applied Medical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging University Clinic and Helmholtz Institute for Biomedical Engineering RWTH – Aachen University Forckenbeckstrasse 5552074AachenGermany
| |
Collapse
|
4
|
Role of Integrins in Modulating Smooth Muscle Cell Plasticity and Vascular Remodeling: From Expression to Therapeutic Implications. Cells 2022; 11:cells11040646. [PMID: 35203297 PMCID: PMC8870356 DOI: 10.3390/cells11040646] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/03/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Smooth muscle cells (SMCs), present in the media layer of blood vessels, are crucial in maintaining vascular homeostasis. Upon vascular injury, SMCs show a high degree of plasticity, undergo a change from a “contractile” to a “synthetic” phenotype, and play an essential role in the pathophysiology of diseases including atherosclerosis and restenosis. Integrins are cell surface receptors, which are involved in cell-to-cell binding and cell-to-extracellular-matrix interactions. By binding to extracellular matrix components, integrins trigger intracellular signaling and regulate several of the SMC function, including proliferation, migration, and phenotypic switching. Although pharmacological approaches, including antibodies and synthetic peptides, have been effectively utilized to target integrins to limit atherosclerosis and restenosis, none has been commercialized yet. A clear understanding of how integrins modulate SMC biology is essential to facilitate the development of integrin-based interventions to combat atherosclerosis and restenosis. Herein, we highlight the importance of integrins in modulating functional properties of SMCs and their implications for vascular pathology.
Collapse
|
5
|
Chakraborty R, Chatterjee P, Dave JM, Ostriker AC, Greif DM, Rzucidlo EM, Martin KA. Targeting smooth muscle cell phenotypic switching in vascular disease. JVS Vasc Sci 2021; 2:79-94. [PMID: 34617061 PMCID: PMC8489222 DOI: 10.1016/j.jvssci.2021.04.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/01/2021] [Indexed: 12/26/2022] Open
Abstract
Objective The phenotypic plasticity of vascular smooth muscle cells (VSMCs) is central to vessel growth and remodeling, but also contributes to cardiovascular pathologies. New technologies including fate mapping, single cell transcriptomics, and genetic and pharmacologic inhibitors have provided fundamental new insights into the biology of VSMC. The goal of this review is to summarize the mechanisms underlying VSMC phenotypic modulation and how these might be targeted for therapeutic benefit. Methods We summarize findings from extensive literature searches to highlight recent discoveries in the mechanisms underlying VSMC phenotypic switching with particular relevance to intimal hyperplasia. PubMed was searched for publications between January 2001 and December 2020. Search terms included VSMCs, restenosis, intimal hyperplasia, phenotypic switching or modulation, and drug-eluting stents. We sought to highlight druggable pathways as well as recent landmark studies in phenotypic modulation. Results Lineage tracing methods have determined that a small number of mature VSMCs dedifferentiate to give rise to oligoclonal lesions in intimal hyperplasia and atherosclerosis. In atherosclerosis and aneurysm, single cell transcriptomics reveal a striking diversity of phenotypes that can arise from these VSMCs. Mechanistic studies continue to identify new pathways that influence VSMC phenotypic plasticity. We review the mechanisms by which the current drug-eluting stent agents prevent restenosis and note remaining challenges in peripheral and diabetic revascularization for which new approaches would be beneficial. We summarize findings on new epigenetic (DNA methylation/TET methylcytosine dioxygenase 2, histone deacetylation, bromodomain proteins), transcriptional (Hippo/Yes-associated protein, peroxisome proliferator-activity receptor-gamma, Notch), and β3-integrin-mediated mechanisms that influence VSMC phenotypic modulation. Pharmacologic and genetic targeting of these pathways with agents including ascorbic acid, histone deacetylase or bromodomain inhibitors, thiazolidinediones, and integrin inhibitors suggests potential therapeutic value in the setting of intimal hyperplasia. Conclusions Understanding the molecular mechanisms that underlie the remarkable plasticity of VSMCs may lead to novel approaches to treat and prevent cardiovascular disease and restenosis.
Collapse
Affiliation(s)
- Raja Chakraborty
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Conn.,Department of Pharmacology, Yale University School of Medicine, New Haven, Conn
| | - Payel Chatterjee
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Conn.,Department of Pharmacology, Yale University School of Medicine, New Haven, Conn
| | - Jui M Dave
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Conn.,Department of Genetics, Yale University School of Medicine, New Haven, Conn
| | - Allison C Ostriker
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Conn.,Department of Pharmacology, Yale University School of Medicine, New Haven, Conn
| | - Daniel M Greif
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Conn.,Department of Genetics, Yale University School of Medicine, New Haven, Conn
| | - Eva M Rzucidlo
- Department Surgery, Section of Vascular Surgery, McLeod Regional Medical Center, Florence, SC
| | - Kathleen A Martin
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Conn.,Department of Pharmacology, Yale University School of Medicine, New Haven, Conn
| |
Collapse
|
6
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
7
|
Anderson HV, Jordan RE, Weisman HF. Concept and Clinical Application of Platelet Glycoprotein IIb/IIIa Inhibition with Abciximab (c7E3 Fab; ReoPro) for the Prevention of Acute Ischemic Syndromes. Clin Appl Thromb Hemost 2016. [DOI: 10.1177/107602969700300407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The platelet membrane glycoprotein (GP) IIb/IIIa integrin receptor is the final common pathway leading to platelet aggregation. Local aggregation commonly occurs following atherosclerotic plaque rupture or other injury to the vascular wall. When GP IIb/IIIa is activated, fibrinogen and von Willebrand factor bind to the receptor with high affinity, crosslinking platelets and locking them to the vessel surface and to each other. This process is central to arterial thrombus formation and consequent acute coronary syndromes, such as myocardial infarction (MI), unstable angina, and abrupt closure following revascularization procedures. Abciximab (c7E3 Fab; ReoPro) is a chimeric monoclonal antibody fragment developed specifically to inhibit GP IIb/IIIa receptor activity and thus prevent platelet aggregation and thrombosis. Abciximab has been evaluated in several clinical studies, the largest of which was the Evaluation of Abciximab for the Prevention of Ischemic Complications (EPIC) trial. This randomized, multicenter, placebo-controlled trial enrolled 2,099 patients at high risk for ischemic complications following coronary revascularization. The patients were randomized into three treatment groups: placebo, abciximab bolus (0.25 mg/kg), or abciximab bolus plus 12-h infusion (10 μg/min). Patients in the abciximab bolus plus infusion group had significant reductions, compared with placebo, in a composite end point of death, nonfatal MI, and urgent coronary intervention within 30 days. These positive, short-term findings were maintained at 6 months of follow-up. Bleeding complications and transfusions were significantly increased in abciximab patients, although there was no increase in bleeding-related death, stroke, or surgery. Retrospective secondary analyses suggested that many of the bleeding events observed in the EPIC trial may have been associated with concomitant high-dose heparin therapy, particularly in lighter weight patients. Subsequent clinical trials have shown that bleeding events can be reduced in patients treated with abciximab by using weight-adjusted heparin dosing without affecting the efficacy of the abciximab bolus plus infusion regimen. Examination of health economic data from the EPIC trial showed that abciximab bolus plus infusion is cost effective as well as clinically beneficial. These results confirm the importance of platelet GP IIb/IIIa receptor blockade in the treatment of acute thrombotic syndromes. Key Words: Platelet aggregation—GP IIb/IIIa receptor—Coronary revascularization— Ischemia.
Collapse
|
8
|
Zhang F, Hao F, An D, Zeng L, Wang Y, Xu X, Cui MZ. The matricellular protein Cyr61 is a key mediator of platelet-derived growth factor-induced cell migration. J Biol Chem 2015; 290:8232-42. [PMID: 25623072 DOI: 10.1074/jbc.m114.623074] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Platelet-derived growth factor (PDGF), a potent chemoattractant, induces cell migration via the MAPK and PI3K/Akt pathways. However, the downstream mediators are still elusive. In particular, the role of extracellular mediators is largely unknown. In this study, we identified the matricellular protein Cyr61, which is de novo synthesized in response to PDGF stimulation, as the key downstream mediator of the ERK and JNK pathways, independent of the p38 MAPK and AKT pathways, and, thereby, it mediates PDGF-induced smooth muscle cell migration but not proliferation. Our results revealed that, when Cyr61 was newly synthesized by PDGF, it was promptly translocated to the extracellular matrix and physically interacted with the plasma membrane integrins α6β1 and αvβ3. We further demonstrate that Cyr61 and integrins are integral components of the PDGF signaling pathway via an "outside-in" signaling route to activate intracellular focal adhesion kinase (FAK), leading to cell migration. Therefore, this study provides the first evidence that the PDGF-induced endogenous extracellular matrix component Cyr61 is a key mediator in modulating cell migration by connecting intracellular PDGF-ERK and JNK signals with integrin/FAK signaling. Therefore, extracellular Cyr61 convergence with growth factor signaling and integrin/FAK signaling is a new concept of growth factor-induced cell migration. The discovered signaling pathway may represent an important therapeutic target in growth factor-mediated cell migration/invasion-related vascular diseases and tumorigenesis.
Collapse
Affiliation(s)
- Fuqiang Zhang
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996 and the Department of Regenerative Medicine, College of Pharmacy, and
| | - Feng Hao
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996 and
| | - Dong An
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996 and College of Life Sciences, Jilin University, Changchun 130021, China
| | - Linlin Zeng
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996 and
| | - Yi Wang
- the Department of Regenerative Medicine, College of Pharmacy, and
| | - Xuemin Xu
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996 and
| | - Mei-Zhen Cui
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996 and
| |
Collapse
|
9
|
Goswami S. Importance of integrin receptors in the field of pharmaceutical & medical science. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abc.2013.32028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
10
|
Cam A, de Mejia EG. RGD-peptide lunasin inhibits Akt-mediated NF-κB activation in human macrophages through interaction with the αVβ3 integrin. Mol Nutr Food Res 2012; 56:1569-81. [PMID: 22945510 DOI: 10.1002/mnfr.201200301] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 06/14/2012] [Accepted: 06/19/2012] [Indexed: 01/22/2023]
Abstract
SCOPE Cardiovascular disease is the leading cause of mortality in the United States and regulation of aberrant macrophage activity under inflammatory conditions is critical for its prevention. The objective was to determine the effect of lunasin on the inhibition of Akt-mediated activation of nuclear factor-kappa B (NF-κB)-dependent markers of inflammation and to characterize the physical interaction of lunasin with the αVβ3 integrin receptor in lipopolysaccharide (LPS)-induced human THP-1 macrophages. METHODS AND RESULTS The effect of lunasin was evaluated in vitro in LPS-induced THP-1 human macrophages using immunoassays, co-immunoprecipitation (Co-IP), and fluorescence confocal microscopy. Lunasin (50 μM) reduced cyclooxygenase-2, inducible nitric oxide synthase, and NO levels by 57.9, 64.5, and 76.2%, respectively, and inhibited the activation of phosphorylated Akt and NF-κB p65 by 59.5 and 74.5%, respectively. Lunasin (50 μM) reduced exogenous release of prostaglandin E(2) and tumor necrosis factor-α by 92.5 and 94.9%, respectively. Vitronectin (10 μg/mL), an integrin ligand, increased expression of proinflammatory markers, whereas lunasin (50 μM) attenuated them. Co-IP of lunasin-treated cells confirmed direct interaction with αVβ3 integrin and LC/MS/MS verified its identity. Lunasin was detected within intracellular vesicles and reduced total αVβ3 intensity as observed by fluorescence microscopy. CONCLUSION Lunasin inhibited αVβ3 integrin-mediated proinflammatory markers and downregulated Akt-mediated NF-κB pathways through interaction with αVβ3 integrin.
Collapse
Affiliation(s)
- Anthony Cam
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 1201 West Gregory Drive, Urbana, IL 61801, USA
| | | |
Collapse
|
11
|
Ouasti S, Kingham PJ, Terenghi G, Tirelli N. The CD44/integrins interplay and the significance of receptor binding and re-presentation in the uptake of RGD-functionalized hyaluronic acid. Biomaterials 2012; 33:1120-34. [DOI: 10.1016/j.biomaterials.2011.10.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 10/06/2011] [Indexed: 11/30/2022]
|
12
|
Impact of polyplex micelles installed with cyclic RGD peptide as ligand on gene delivery to vascular lesions. Gene Ther 2011; 19:61-9. [DOI: 10.1038/gt.2011.74] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
13
|
Luo LL, Wang GX, Li YL, Yin TY, Jiang T, Ruan CG. Layer-by-layer assembly of chitosan and platelet monoclonal antibody to improve biocompatibility and release character of PLLA coated stent. J Biomed Mater Res A 2011; 97:423-32. [DOI: 10.1002/jbm.a.33066] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 11/19/2010] [Accepted: 01/04/2011] [Indexed: 11/12/2022]
|
14
|
Osherov AB, Gotha L, Cheema AN, Qiang B, Strauss BH. Proteins mediating collagen biosynthesis and accumulation in arterial repair: novel targets for anti-restenosis therapy. Cardiovasc Res 2011; 91:16-26. [PMID: 21245059 DOI: 10.1093/cvr/cvr012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Events contributing to restenosis after coronary interventions include platelet aggregation, inflammatory cell infiltration, growth factor release, and accumulation of smooth muscle cells (SMCs) and extracellular matrix (ECM). The ECM is composed of various collagen subtypes and proteoglycans and over time constitutes the major component of the mature restenotic plaque. The pathophysiology of collagen accumulation in the ECM during arterial restenosis is reviewed. Factors regulating collagen synthesis and degradation, including various cytokines and growth factors involved in the process, may be targets for therapies aimed at prevention of in-stent restenosis.
Collapse
Affiliation(s)
- Azriel B Osherov
- Schulich Heart Program, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Room A-253, Toronto, Ontario, Canada M4N 3M5
| | | | | | | | | |
Collapse
|
15
|
Schulz S, Sibbing D, Braun S, Morath T, Mehilli J, Massberg S, Byrne RA, Schömig A, Kastrati A. Platelet response to clopidogrel and restenosis in patients treated predominantly with drug-eluting stents. Am Heart J 2010; 160:355-61. [PMID: 20691843 DOI: 10.1016/j.ahj.2010.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 05/06/2010] [Indexed: 02/02/2023]
Abstract
BACKGROUND Preclinical studies suggest a relationship between early thrombotic response after vascular injury and later development of restenosis. The aim of this study was to assess the impact of platelet response to clopidogrel on the risk of restenosis after drug-eluting stenting (DES). METHODS A total of 1,608 consecutive patients were previously enrolled in a study on the relation between platelet reactivity and outcomes after DES. All patients received a loading dose of 600 mg clopidogrel. Blood samples for the assessment of adenosine diphosphate-induced platelet aggregation with multiple electrode platelet aggregometry were drawn directly before percutaneous coronary intervention. Clopidogrel low response was defined as upper quintile of multiple electrode platelet aggregometry measurements. Accordingly, 323 patients (20%) were considered as low and 1,285 (80%) as normal responders. Primary end point of the present study was target lesion revascularization at 1 year. Secondary end points included binary angiographic restenosis and late lumen loss at 6- to 8-month angiography. RESULTS Target lesion revascularization rates were comparable in both groups (10.9% vs 9.5%, hazard rate [HR] 1.2, 95% CI 0.8-1.7, P = .441). Follow-up angiography revealed no difference in binary angiographic restenosis (13.9% vs 15.9%, P = .445) and late lumen loss (0.32 +/- 0.64 vs 0.35 +/- 0.63 mm, P = .477). Low responders had significantly more stent thromboses (2.5% vs 0.5%, HR 5.4, 95% CI 1.9-15.6, P = .002), Q wave myocardial infarctions (2.5% vs 0.6%, HR 4.0, 95% CI 1.5-10.7, P = .005), and ischemic strokes (1.3% vs 0.2%, HR 5.4, 95% CI 1.2-24.0, P = .028) at 1 year. CONCLUSION Low platelet responsiveness to clopidogrel, a known predictor of thrombotic complications, does not have a significant impact on restenosis after DES.
Collapse
|
16
|
Increased static pressure promotes migration of vascular smooth muscle cells: involvement of the Rho-kinase pathway. J Cardiovasc Pharmacol 2008; 51:55-61. [PMID: 18209569 DOI: 10.1097/fjc.0b013e31815b9d26] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vascular smooth muscle cell (VSMC) migration plays a pivotal role in the pathogenesis of arteriosclerosis, under influences of various mechanical factors. Thus, we examined whether static pressure promotes VSMC migration and if so, whether Rho-kinase is involved. Rat VSMCs were cultured on chambers coated on fibronectin, vitronectin, laminin, or type IV collagen, under pressure-free conditions and at 90 and 180 mm Hg. In monolayer-wounding assay, VSMC migration was significantly increased after 72 hours at 180 mm Hg on both fibronectin (11.3 +/- 3.4-fold vs. pressure-free conditions) and vitronectin (10.6 +/- 0.7-fold; both P < 0.05). In Boyden chamber assay, the VSMC migration was again significantly increased at 180 mm Hg on both fibronectin (4.0 +/- 0.5-fold) and vitronectin (5.0 +/- 0.8-fold; both P < 0.05). Neutralizing antibodies against beta1-, beta3- and beta5-integrins, all of which play an important role in cell migration, significantly inhibited the pressure-promoted VSMC migration. Static pressure also significantly increased Rho-kinase activity in VSMC, as evaluated by the extent of phosphorylation of its downstream substrate, ezrin-radixin-moesin. Fasudil, a selective Rho-kinase inhibitor, significantly suppressed the pressure-promoted VSMC migration with reduced Rho-kinase activity. These results indicate that increased static pressure promotes VSMC migration through the integrin/Rho-kinase signaling, suggesting the therapeutic importance of this mechanism for the treatment of hypertensive vascular diseases.
Collapse
|
17
|
Chen Y, Cheng D, Marugan JJ, Manthey C, Tomczuk B, Huebert N. Pharmacokinetics of TDP223206 following intravenous and oral administration to intact rats and intravenous administration to bile duct-cannulated rats. Biopharm Drug Dispos 2008; 29:219-30. [PMID: 18260095 DOI: 10.1002/bdd.601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The pharmacokinetics of TDP223206 was studied following single intravenous and oral administrations in rats. A mixture of TDP223206 and (14)C-TDP223206 were administered to intact and bile duct-cannulated rats. Following intravenous administration, plasma concentrations declined biphasically. The AUC(inf) increased linearly with dose but was not dose proportional. The PK parameters of TDP223206 indicated low clearance (254-386 ml/h/kg) and a moderate volume of distribution (968-1883 ml/kg). The bioavailability was 32.95% and 24.46% for 10 and 50 mg/kg oral doses, respectively. (14)C-TDP223206 was distributed widely into different tissues with small intestine, liver, kidneys and large intestine having large tissue to plasma ratios. (14)C-TDP223206 was the major circulating component in the plasma. A total of 91.2% of administered radioactivity of (14)C-TDP223206 was recovered in bile indicating that biliary excretion was the major pathway for drug elimination. (14)C-TDP223206-acyl glucuronides were the major metabolites in bile. The oxo-(14)C-TDP223206 was the major metabolite in plasma and an important metabolite in bile. Two forms of diastereomeric acyl glucuronides of (14)C-TDP223206 were detected in bile with similar LC/MS intensities suggesting a similar biotransformation capacity. Only one form of these (14)C-TDP223206-acyl glucuronides was detected in plasma suggesting that enterohepatic recirculation was related to the nature of the stereo-isomers.
Collapse
Affiliation(s)
- Yanmin Chen
- Johnson & Johnson Pharmaceutical Research and Development LLC, Welsh and McKean Roads, Spring House, PA 19477, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Kokubo T, Uchida H, Choi ET. Integrin alpha(v)beta(3) as a target in the prevention of neointimal hyperplasia. J Vasc Surg 2007; 45 Suppl A:A33-8. [PMID: 17544022 PMCID: PMC1939972 DOI: 10.1016/j.jvs.2007.02.069] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 02/21/2007] [Indexed: 11/17/2022]
Abstract
Although major advances have been made in the prevention and treatment of restenosis following coronary and peripheral interventions, the persistent complications of thrombosis and reintervention remain a mainstay for repeat hospitalizations in this patient population. For many years, a ubiquitous cell surface receptor called alpha(v)beta(3) integrin was the target of investigators in the prevention of restenosis because its interaction with the extracellular matrix was believed to coordinate the migration of smooth muscle cells (SMCs) from the media to the intima, the seminal event in the formation of intimal occlusive lesion. After the publication of uniformly positive animal studies demonstrating that alpha(v)beta(3) integrin blockade led to a significant reduction in new intimal (neointimal) lesion formation, early clinical trials supported the association of avoidance of target lesion revascularization and the use of antagonists to the SMC integrin alpha(v)beta(3) and its related platelet integrin alpha(IIb)beta(3). However, a series of clinical trials subsequently demonstrated that these antagonists did not necessarily prevent revascularizations by inhibiting intimal hyperplasia per se. Additional animal studies subsequently showed that, indeed, in the setting of pre-existing SMCs in the intimal lesion (ie, atherosclerotic plaque, fatty streaks), inhibiting SMC migration by way of beta(3) integrin blockade was an ineffective approach in the prevention of intimal hyperplasia and restenosis. However, given the wealth of basic and clinical information on the alpha(v)beta(3) integrin and its antagonists, we discuss in this article our new approach to this old solution by targeting a new clinical problem of early failure arteriovenous access for hemodialysis. Given the uniqueness of arteriovenous access in that there are essentially no significant atherosclerotic lesions in the artery and vein prior to the anastomosis, the seminal event of the migration of SMCs from the media to the neointima could by targeted once again with beta(3) integrin antagonists.
Collapse
Affiliation(s)
- Taku Kokubo
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
19
|
Frame MD, Rivers RJ, Altland O, Cameron S. Mechanisms initiating integrin-stimulated flow recruitment in arteriolar networks. J Appl Physiol (1985) 2007; 102:2279-87. [PMID: 17379749 DOI: 10.1152/japplphysiol.00537.2006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Our purpose was to investigate the local mechanisms involved in network-wide flow and diameter changes observed with localized downstream vitronectin receptor ligation; we tested specific K or Cl channels known to be involved in either dilation or elevated permeability following vitronectin receptor activation and tested integrin-linked pathway elements of tyrosine phosphorylation and protein kinase C (PKC). Arteriolar networks were observed in the cheek pouch tissue of anesthetized (pentobarbital sodium, 70 mg/kg) hamsters ( n = 86) using intravital microscopy. Terminal arteriolar branches of the networks were stimulated with micropipette LM609 (0.5–10 μg/ml, 60 s) alone or with inhibitors (separate micropipette). Hemodynamic changes (diameter, red blood cell flux, velocity) were observed at the upstream entrance to the network. LM609 alone stimulated first an increase in wall shear stress (WSS), followed by a dilation that recovered WSS to baseline or below. K channel inhibition (glybenclamide, 4-AP) had no effect on the initial peak in WSS, but decreased remote vasodilation. Cl channel inhibition (DIDS, IAA-94, niflumic acid) or inhibition of PKC (chelerythrine) prevented the initial peak in WSS and decreased remote vasodilation. Inhibition of tyrosine phosphorylation (genistein) prevented both. With the use of nitro-arginine at the observation site, the initial peak in WSS was not affected, but remote vasodilation was decreased. We conclude the remote response consists of an initial peak in WSS that relies on both PKC activity and depolarization downstream, leading to an upstream flow mediated dilation and a secondary remote dilation that relies on hyperpolarization downstream at the stimulus site; both components require tyrosine phosphorylation downstream.
Collapse
Affiliation(s)
- Mary D Frame
- Department of Biomedical Engineering, Stony Brook University, HSC T18-030, Stony Brook, NY 11794-8181, USA.
| | | | | | | |
Collapse
|
20
|
Zhou Y, Peng H, Ji Q, Qi J, Zhu Z, Yang C. Discovery of small molecule inhibitors of integrin αvβ3 through structure-based virtual screening. Bioorg Med Chem Lett 2006; 16:5878-82. [PMID: 16982193 DOI: 10.1016/j.bmcl.2006.08.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Revised: 07/28/2006] [Accepted: 08/11/2006] [Indexed: 11/15/2022]
Abstract
Inhibitors of integrin alphavbeta3 have been implicated in the treatment of a variety of diseases, including tumor metastasis, neovascularization, osteoporosis, and rheumatoid arthritis. It is therefore desirable to develop new types of small molecule inhibitors of integrin alphavbeta3. Here we describe the discovery of novel classes of small molecule inhibitors, via structure-based virtual screening, that target the ligand binding site of integrin alphavbeta3. Application of the docking procedure for screening of a commercially available compound database resulted in a 1774-fold reduction in the size of the screening set (88695 to 50 compounds) and gave a hit-rate of 14% upon biological evaluation (IC50 value ranging from 30 to 200 microM). The best hit, compound 37, 3,4-dichloro-phenylbiguanide, showed inhibitory activity, in a time- and dose-dependent manner, in both cell motility and angiogenesis assays. Based on the best hit, compound 37, a more effective derivative compound 62 has been identified. Furthermore, molecular graphics analyses of a series of substituted phenylbiguanides were carried out to predict the binding mode between the active compounds and integrin alphavbeta3. Our results indicate that the substituted phenylbiguanides might be involved in the inhibition of bivalent cation-mediated ligand binding of integrin alphavbeta3.
Collapse
Affiliation(s)
- Yuan Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, PR China
| | | | | | | | | | | |
Collapse
|
21
|
Spatial Screening for the Identification of the Bioactive Conformation of Integrin Ligands. Top Curr Chem (Cham) 2006. [DOI: 10.1007/128_052] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
22
|
Voisard R, Alan M, Müller LV, Baur R, Hombach V. Effects of abciximab on key pattern of human coronary restenosis in vitro: impact of the SI/MPL-ratio. BMC Cardiovasc Disord 2006; 6:14. [PMID: 16595000 PMCID: PMC1475639 DOI: 10.1186/1471-2261-6-14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 04/04/2006] [Indexed: 11/10/2022] Open
Abstract
Background The significant reduction of angiographic restenosis rates in the ISAR-SWEET study (intracoronary stenting and antithrombotic regimen: is abciximab a superior way to eliminate elevated thrombotic risk in diabetes) raises the question of whether abciximab acts on clopidogrel-independent mechanisms in suppressing neointimal hyperplasia. The current study investigates the direct effect of abciximab on ICAM-1 expression, migration and proliferation. Methods ICAM-1: Part I of the study investigates in cytoflow studies the effect of abciximab (0.0002, 0.002, 0.02, 0.2, 2.0, and 20.0 μg/ml) on TNF-α induced expression of intercellular adhesion molecule 1 (ICAM-1). Migration: Part II of the study explored the effect of abciximab (0.0002, 0.002, 0.02, 0.2, 2.0, and 20.0 μg/ml) on migration of HCMSMC over a period of 24 h. Proliferation: Part III of the study investigated the effect of abciximab (0.0002, 0.002, 0.02, 0.2, 2.0, and 20.0 μg/ml) on proliferation of HUVEC, HCAEC, and HCMSMC after an incubation period of 5 days. Results ICAM-1: In human venous endothelial cells (HUVEC), human coronary endothelial cells (HCAEC) and human coronary medial smooth muscle cells (HCMSMC) no inhibitory or stimulatory effect on expression of ICAM-1 was detected. Migration: After incubation of HCMSMC with abciximab in concentrations of 0.0002 – 2 μg/ml a stimulatory effect on cell migration was detected, statistical significance was achieved after incubation with 0.002 μg/ml (p < 0.05), 0.002 μg/ml (p < 0.001), and 0.2 μg/ml (p < 0.05). Proliferation: Small but statistically significant antiproliferative effects of abciximab were detected after incubation of HUVEC (0.02 and 2.0 μg/ml; p = 0.01 and p < 0.01), HCAEC (2.0 and 20.0 μg/ml; p < 0.05 and p < 0,01), and HCMSMC (2.0 and 20.0 μg/ml; p < 0.05 and p < 0.05). The significant inhibition (SI) of cell proliferation found in HCAEC and HCMSMC was achieved with drug concentrations more than 10 times beyond the maximal plasma level (MPL), resulting in a SI/MPL-ratio > 1. Conclusion Thus, the anti-restenotic effects of systemically administered abciximab reported in the ISAR-SWEET-study were not caused by a direct inhibitory effect on ICAM-1 expression, migration or proliferation.
Collapse
Affiliation(s)
- Rainer Voisard
- Department of Internal Medicine II – Cardiology, University of Ulm, Robert-Kochstrasse 8, D-89081 Ulm, (Rainer Voisard, M.D., Mustafa Alan, Regine Baur, Vinzenz Hombach, M.D.), Germany
| | - Mustafa Alan
- Department of Internal Medicine II – Cardiology, University of Ulm, Robert-Kochstrasse 8, D-89081 Ulm, (Rainer Voisard, M.D., Mustafa Alan, Regine Baur, Vinzenz Hombach, M.D.), Germany
| | - Lutz von Müller
- Department of Virology, Institute of Mikrobiology and Immunology, University of Ulm, Robert-Kochstrasse 8, D-89081 Ulm, (Lutz von Müller, M.D.), Germany
| | - Regine Baur
- Department of Internal Medicine II – Cardiology, University of Ulm, Robert-Kochstrasse 8, D-89081 Ulm, (Rainer Voisard, M.D., Mustafa Alan, Regine Baur, Vinzenz Hombach, M.D.), Germany
| | - Vinzenz Hombach
- Department of Internal Medicine II – Cardiology, University of Ulm, Robert-Kochstrasse 8, D-89081 Ulm, (Rainer Voisard, M.D., Mustafa Alan, Regine Baur, Vinzenz Hombach, M.D.), Germany
| |
Collapse
|
23
|
Asano Y, Ihn H, Yamane K, Jinnin M, Mimura Y, Tamaki K. Increased expression of integrin alpha(v)beta3 contributes to the establishment of autocrine TGF-beta signaling in scleroderma fibroblasts. THE JOURNAL OF IMMUNOLOGY 2006; 175:7708-18. [PMID: 16301681 DOI: 10.4049/jimmunol.175.11.7708] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The constitutive secretion of latent TGF-beta by many cell types in culture suggests that extracellular mechanisms to control the activity of this potent cytokine are important in the pathogenesis of the diseases in which this cytokine may be involved, including fibrotic disorders. In this study, we focused on the alpha(v)beta3 integrin, which is recently demonstrated to function as an active receptor for latent TGF-beta1 through its interaction with latency-associated peptide-beta1, and investigated the involvement of this integrin in the pathogenesis of scleroderma. Scleroderma fibroblasts exhibited increased alpha(v)beta3 expression compared with normal fibroblasts in vivo and in vitro. In scleroderma fibroblasts, ERK pathway was constitutively activated and such abnormality induced the up-regulation of alpha(v)beta3. Transient overexpression of alpha(v)beta3 in normal fibroblasts induced the increase in the promoter activity of human alpha2(I) collagen gene and the decrease in that of human MMP-1 gene. These effects of alpha(v)beta3 were almost completely abolished by the treatment with anti-TGF-beta Ab or TGF-beta1 antisense oligonucleotide. Furthermore, the addition of anti-alpha(v)beta3) Ab reversed the expression of type I procollagen protein and MMP-1 protein, the promoter activity of human alpha2(I) collagen gene, and the myofibroblastic phenotype in scleroderma fibroblasts. These results suggest that the up-regulated expression of alpha(v)beta3 contributes to the establishment of autocrine TGF-beta loop in scleroderma fibroblasts, and this integrin is a potent target for the treatment of scleroderma.
Collapse
Affiliation(s)
- Yoshihide Asano
- Department of Dermatology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Graef T, Steidl U, Nedbal W, Rohr U, Fenk R, Haas R, Kronenwett R. Use of RNA interference to inhibit integrin subunit alphaV-mediated angiogenesis. Angiogenesis 2006; 8:361-72. [PMID: 16400518 DOI: 10.1007/s10456-005-9026-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Revised: 11/14/2005] [Accepted: 12/02/2005] [Indexed: 11/25/2022]
Abstract
One of the central molecules in capillary formation during angiogenesis is the integrin alphaVbeta3. The aim of this study was to inhibit alphaV-mediated angiogenesis in vitro using RNAs (siRNA) as well as antisense oligodeoxyribonucleotides (asON). Five siRNAs, against the alphaV chain of alphaVbeta3, and three asON, which had the respective sequence of the antisense sequence of three of the siRNAs molecules, were examined. Two of the siRNAs and their respective asON were designed on the basis of computer-predicted secondary structure analysis of alphaV mRNA. The different molecules were transfected into human umbilical vein endothelials cells (HUVEC) using lipofection. Following stimulation by PMA, two siRNAs showed a dose-dependent inhibition of PMA-induced alphaV mRNA and protein upregulation, as assessed by real-time RT-PCR and flow cytometry. At a concentration of 25 nM a complete inhibition of protein upregulation was found using siRNAs while transfection of the respective asON sequences reduced the protein upregulation only by 44%. To evaluate the anti-angiogenic potential a cell culture model of human angiogenesis based on the co-cultivation of endothelial cells and dermal fibroblasts was used. Transfection of the siRNA sequence (50 nM) resulted in an inhibition of the total length of capillary-like tubules by 40.6% in comparison to 21.1% using the respective asON sequence. In conclusion, siRNA-based downregulation of alphaV expression showed a stronger inhibition of capillary tube formation in an angiogenesis in vitro assay, than asON having the same sequence as the antisense strand of the siRNAs. Therefore, siRNAs are useful tools for functional alphaV knock-down experiments and might be a therapeutic alternative for antagonists which bind directly to the integrins alphaVbeta3 or alphaVbeta5.
Collapse
Affiliation(s)
- Thorsten Graef
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Germany.
| | | | | | | | | | | | | |
Collapse
|
25
|
Choi ET, Collins ET, Marine LA, Uberti MG, Uchida H, Leidenfrost JE, Khan MF, Boc KP, Abendschein DR, Parks WC. Matrix Metalloproteinase-9 Modulation by Resident Arterial Cells Is Responsible for Injury-Induced Accelerated Atherosclerotic Plaque Development in Apolipoprotein E–Deficient Mice. Arterioscler Thromb Vasc Biol 2005; 25:1020-5. [PMID: 15746435 DOI: 10.1161/01.atv.0000161275.82687.f6] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Although matrix metalloproteinase-9 (MMP-9) has been implicated in atherosclerotic plaque instability, the exact role it plays in the plaque development and progression remains largely unknown. We generated apolipoprotein E (apoE)-deficient (apoE-/-) MMP-9-deficient (MMP-9-/-) mice to determine the mechanisms and the main cell source of MMP-9 responsible for the plaque composition during accelerated atherosclerotic plaque formation. METHODS AND RESULTS Three weeks after temporary carotid artery ligation revealed that while on a Western-type diet, apoE-/- MMP-9-/- mice had a significant reduction in intimal plaque length and volume compared with apoE-/- MMP-9+/+ mice. The reduction in plaque volume correlated with a significantly lower number of intraplaque cells of resident cells and bone marrow-derived cells. To determine the cellular origin of MMP-9 in plaque development, bone marrow transplantation after total-body irradiation was performed with apoE-/- MMP-9+/+ and apoE-/- MMP-9-/- mice, which showed that only MMP-9 derived from resident arterial cells is required for plaque development. CONCLUSIONS MMP-9 is derived from resident arterial cells and is required for early atherosclerotic plaque development and cellular accumulation in apoE-/- mice.
Collapse
Affiliation(s)
- Eric T Choi
- Department of Surgery, Washington University School of Medicine, St. Louis, Mo, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Linde J, Strauss BH. Pharmacological treatment for prevention of restenosis. Expert Opin Emerg Drugs 2005; 6:281-302. [PMID: 15989527 DOI: 10.1517/14728214.6.2.281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Coronary artery disease (CAD) is the leading cause of mortality and morbidity among adults in the Western world. Coronary artery bypass grafting and percutaneous coronary interventions (PCI) have gained widespread acceptance for the treatment of symptomatic CAD. There has been an explosive growth worldwide in the utilisation of PCI, such as balloon angioplasty and stenting, which now accounts for over 50% of coronary revascularisation. Despite the popularity of PCI, the problem of recurrent narrowing of the dilated artery (restenosis) continues to vex investigators. In recent years, significant advances have occurred in the understanding of restenosis. Two processes seem to contribute to restenosis: remodelling (vessel size changes) and intimal hyperplasia (vascular smooth muscle cell [VSMC] proliferation and extracellular matrix [ECM] deposition). Despite considerable efforts, pharmacological approaches to decrease restenosis have been largely unsuccessful and the only currently applied modality to reduce the restenosis rate is stenting. However, stenting only prevents remodelling and does not inhibit intimal hyperplasia. Several potential targets for inhibiting restenosis are currently under investigation including platelet activation, the coagulation cascade, VSMC proliferation and migration, and ECM synthesis. In addition, new approaches for local drug therapy, such as drug eluting stents, are currently being evaluated in preclinical and clinical studies. In this article, we critically review the current status of drugs that are being evaluated for restenosis at various stages of development (in vitro, preclinical animal models and human trials).
Collapse
Affiliation(s)
- J Linde
- The Roy and Ann Foss Interventional Cardiology Research Program, Terrence Donnelly Heart Center, 30 Bond Street, St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
| | | |
Collapse
|
27
|
Manoharan G, Maynard SJ, Adgey AA. The therapeutic use of glycoprotein IIb/IIIa inhibitors in acute coronary syndromes. Expert Opin Investig Drugs 2005; 8:555-66. [PMID: 15992115 DOI: 10.1517/13543784.8.5.555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Acute coronary syndrome (ACS) embraces the clinical diagnoses of unstable angina and non-Q-wave myocardial infarction (NON-Q-MI). Conventional treatment for these conditions with aspirin, heparin and other anti-anginal drugs has its limitations. Treatment of acute symptoms with lytic therapy has not been beneficial. A large proportion of patients in this high-risk group will eventually need revascularisation due to ongoing symptoms. Platelet aggregation plays a crucial role in the pathogenesis of ACS and ischaemic complications of coronary intervention. The glycoprotein (GP) IIb/IIIa receptor, which belongs to a family of surface receptors called integrins, is the final step in this pathway. This has led to the development and use of GP IIb/IIIa inhibitors as potential therapeutic agents. Inhibitors developed so far include abciximab, a chimeric monoclonal antibody, and more recently, novel synthetic intravenous and orally administered competitive integrin blocking agents. To date, abciximab, tirofiban and eptifibatide are commercially available for clinical use. Completed clinical trials have looked at these agents as a primary treatment and as an adjunct to intervention for ACS; they have shown improvement over existing treatments, with reductions in the incidence of death, myocardial infarction (MI), refractory ischaemia and need for urgent revascularisation. Risk of bleeding and thrombocytopenia is low, and appears to be dose related and associated with concomitant treatment with high dose heparin. Cost benefit has been proven.
Collapse
Affiliation(s)
- G Manoharan
- Regional Medical Cardiology Centre, Royal Victoria Hospital, Grosvenor Road, Belfast BT12 6BA, UK
| | | | | |
Collapse
|
28
|
Cabrera-Serrano A, García-Arrarás JE. RGD-containing peptides inhibit intestinal regeneration in the sea cucumber Holothuria glaberrima. Dev Dyn 2005; 231:171-8. [PMID: 15305297 DOI: 10.1002/dvdy.20112] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The sea cucumber Holothuria glaberrima is an echinoderm capable of regenerating its viscera. Previous studies from our group have shown a striking remodeling of the extracellular matrix (ECM) during intestinal regeneration. To study the role of the ECM during regeneration, we have focused on the RGD sequences present in many ECM molecules. Regenerating animals were treated with an RGDS (Arg-Gly-Asp-Ser) peptide that competes with the interaction between RGD sequence and cellular integrins. Saline and RGES (Arg-Gly-Glu-Ser) peptide injections were done as controls. The size of the regenerating intestine was determined, and the regenerating structures were analyzed by immunohistochemistry for the presence of collagen and fibronectin, as well as for muscle and other cells. The results show a delay in intestinal regeneration in animals injected with the RGDS peptide, suggesting that the ECM-integrin interaction plays an important function in the regenerative process.
Collapse
|
29
|
Sadeghi MM, Krassilnikova S, Zhang J, Gharaei AA, Fassaei HR, Esmailzadeh L, Kooshkabadi A, Edwards S, Yalamanchili P, Harris TD, Sinusas AJ, Zaret BL, Bender JR. Detection of injury-induced vascular remodeling by targeting activated alphavbeta3 integrin in vivo. Circulation 2004; 110:84-90. [PMID: 15210600 DOI: 10.1161/01.cir.0000133319.84326.70] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The alpha(v)beta3 integrin plays a critical role in cell proliferation and migration. We hypothesized that vascular cell proliferation, a hallmark of injury-induced remodeling, can be tracked by targeting alpha(v)beta3 integrin expression in vivo. METHODS AND RESULTS RP748, a novel 111In-labeled alpha(v)beta3-specific radiotracer, was evaluated for its cell-binding characteristics and ability to track injury-induced vascular proliferation in vivo. Three groups of experiments were performed. In cultured endothelial cells (ECs), TA145, a cy3-labeled homologue of RP748, localized to alpha(v)beta3 at focal contacts. Activation of alpha(v)beta3 by Mn2+ led to increased EC binding of TA145. Left common carotid artery wire injury in apolipoprotein E-/- mice led to vascular wall expansion over a period of 4 weeks. RP748 (7.4 MBq) was injected into groups of 9 mice at 1, 3, or 4 weeks after left carotid injury, and carotids were harvested for autoradiography. Relative autographic intensity, defined as counts/pixel of the injured left carotid area divided by counts/pixel of the uninjured right carotid area, was higher at 1 and 3 weeks (1.8+/-0.1 and 1.9+/-0.2, respectively) and decreased significantly by 4 weeks after injury (1.4+/-0.1, P<0.05). Carotid alpha(v) and beta3 integrin expression was maximal at 1 week and decreased by 4 weeks after injury. The proliferation index, as determined by Ki67 staining, followed a temporal pattern similar to that of RP748 uptake. Dynamic gamma imaging demonstrated rapid renal clearance of RP748. CONCLUSIONS RP748 has preferential binding to activated alpha(v)beta3 integrin and can track the injury-induced vascular proliferative process in vivo.
Collapse
Affiliation(s)
- Mehran M Sadeghi
- Raymond and Beverly Sackler Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Conn, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Choi ET, Khan MF, Leidenfrost JE, Collins ET, Boc KP, Villa BR, Novack DV, Parks WC, Abendschein DR. Beta3-integrin mediates smooth muscle cell accumulation in neointima after carotid ligation in mice. Circulation 2004; 109:1564-9. [PMID: 15007005 DOI: 10.1161/01.cir.0000121733.68724.ff] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pharmacological blockade of beta3-integrins inhibits neointimal lesion formation in nonmouse animal models of arterial injury. In contrast, beta3-integrin-deficient (beta3-/-) mice are not protected from neointimal lesion formation after arterial injury. We investigated this discrepancy in beta3-/- and wild-type (beta3+/+) mice using different models of injury. METHODS AND RESULTS After disruption of the carotid with a transluminal probe, there was no significant difference in neointimal thickening between beta3-/- and beta3+/+ mice. However, after ligation of the carotid without medial disruption, there was reduced neointimal thickening in beta3-/- mice compared with beta3+/+ mice at intervals up to 3 months. Lesion reduction in beta3-/- mice was associated with fewer intimal smooth muscle cells (SMCs) without a difference in SMC apoptosis or proliferation rate compared with beta3+/+ mice, consistent with reduced SMC migration from the media into the intima of beta3-/- mice. Moreover, combined eccentric medial disruption and ligation of the carotid in beta3-/- mice resulted in neointimal lesion formation only at the site of medial disruption. Transplantation of bone marrow cells harvested from beta3+/+ mice into irradiated beta3-/- mice resulted in reduced neointimal lesion formation after carotid ligation injury, confirming the importance of alpha(v)beta3 and not alpha(IIb)beta3 in the attenuated response. CONCLUSIONS The alpha(v)beta3-integrin mediates intimal SMC accumulation that contributes to neointimal thickening in the setting of arterial ligation.
Collapse
Affiliation(s)
- Eric T Choi
- Department of Surgery, Washington University School of Medicine, St Louis, Mo, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gabeler EEE, van Hillegersberg R, Sluiter W, Kliffen M, Statius van Eps RG, Honkoop J, Carlier SG, van Urk H. Arterial wall strength after endovascular photodynamic therapy. Lasers Surg Med 2003; 33:8-15. [PMID: 12866116 DOI: 10.1002/lsm.10187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Vascular photodynamic therapy (PDT) inhibits intimal hyperplasia (IH) induced by angioplasty in rat iliac arteries by eradicating the proliferating smooth muscle cells. This process may jeopardise the structure and strength of the arterial wall, reflected by a decreased bursting pressure. STUDY DESIGN/MATERIALS AND METHODS Thirty male Wistar rats of 250-300 g were subdivided into 3 groups (n = 10). In all groups, IH was induced by balloon injury (BI). One experimental group received PDT at 50 J/cm diffuser length, the other group at 100 J/cm diffuser length. The third group served as control group and received no PDT. In half of each group the bursting pressure was analyzed after 2 hours (n = 5), in the other half after 1 year. RESULTS Two hours after the procedure the bursting pressure was 3.37 +/- 0.58 (+/-SEM) bar in the BI + PDT 50 and 3.96 +/- 0.43 bar in the BI + PDT 100 group, compared to 2.20 +/- 0.27 bar in the BI group (P < 0.05). After 1 year these values were 3.18 +/- 0.87 bar in the BI + PDT 50 (P < 0.05) and 2.02 +/- 0.31 bar in the BI + PDT 100 group, compared to 2.10 +/- 0.30 bar in the BI group (NS). In the BI + PDT 100 group, 3 out of 5 rats appeared to have aneurysmal dilatation after 1 year. CONCLUSIONS Endovascular PDT increases the arterial wall strength as measured by the bursting pressure at short-term. After 1 year, wall strength is not diminished as measured by bursting pressure, but aneurysmal dilatation nevertheless developed with 100 J/cm. dl. This may limit the use of high energy PDT.
Collapse
|
32
|
Sajid M, Zhao R, Pathak A, Smyth SS, Stouffer GA. Alphavbeta3-integrin antagonists inhibit thrombin-induced proliferation and focal adhesion formation in smooth muscle cells. Am J Physiol Cell Physiol 2003; 285:C1330-8. [PMID: 12878490 DOI: 10.1152/ajpcell.00475.2002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Alphavbeta3-integrin antagonists reduced neointimal formation following vascular injury in eight different animal models. Because alpha-thrombin contributes to neointimal formation, we examined the hypothesis that alphavbeta3-integrins influence alpha-thrombin-induced signaling. Cultured rat aortic smooth muscle cells (RASMC) expressed alphavbeta3-integrins as demonstrated by immunofluorescence microscopy and fluorescence-activated cell sorting analysis. Proliferative responses to alpha-thrombin were partially inhibited by anti-beta3-integrin monoclonal antibody F11 and by cyclic RGD peptides. Immunofluorescence microscopy showed that alpha-thrombin stimulated a rapid increase in the formation of focal adhesions as identified by vinculin staining and that this effect was partially inhibited by alphavbeta3 antagonists. Beta3-integrin staining was diffuse in quiescent RASMC and did not concentrate at sites of focal adhesions following thrombin treatment. Alpha-thrombin elicited a time-dependent increase in activation of c-Jun NH2-terminal kinase-1 (JNK1) and in tyrosine phosphorylation of focal adhesion kinase (FAK). Alphavbeta3-integrin antagonists partially inhibited increases in JNK1 activity but had no effect on FAK phosphorylation. In SMC isolated from beta3-integrin-deficient mice, focal adhesion formation was impaired in response to thrombin but not sphingosine-1-phosphate, a potent activator of Rho. In summary, alphavbeta3-integrins play an important role in alpha-thrombin-induced proliferation and focal adhesion formation in RASMC.
Collapse
Affiliation(s)
- M Sajid
- Division of Cardiology, Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599-7075, USA
| | | | | | | | | |
Collapse
|
33
|
Michel JB. Anoikis in the cardiovascular system: known and unknown extracellular mediators. Arterioscler Thromb Vasc Biol 2003; 23:2146-54. [PMID: 14551156 DOI: 10.1161/01.atv.0000099882.52647.e4] [Citation(s) in RCA: 179] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Anoïkis is defined as programmed cell death induced by the loss of cell/matrix interactions. Adhesion to structural glycoproteins of the extracellular matrix is necessary for survival of the differentiated adherent cells in the cardiovascular system, including endothelial cells, smooth muscle cells, fibroblasts, and cardiac myocytes. Adhesion is also a key factor for the differentiation of mesenchymal stem cells. In particular, fibronectin is considered a factor of survival and differentiation for many adherent cells. Adhesion generates cell tensional integrity (tensegrity) and repression of apoptotic signals, whereas detachment has the opposite effect. Anoïkis plays a physiological role by regulating cell homeostasis in tissues. However, anoïkis can also be involved in pathological processes, as illustrated by the resistance to anoïkis in cancer and its enhancement in degenerative tissue remodeling. Extracellular mediators of anoïkis include matrix retraction, leading to loss of tensegrity in fibroblasts, pharmacological disengagement of integrins by RGD-like peptides and fragments of fibronectin, and focal adhesion disassembly by fragments of thrombospondin, plasminogen activator-1, and high-molecular-weight kininogen. In addition to binding of the RGD peptide by integrins, the engagement of the heparin binding sites of adhesive glycoproteins with glycosaminoglycans on the cell surface is also involved in the prevention of cell detachment-induced apoptosis. Proteases able to degrade adhesive glycoproteins, such as fibronectin, induce anoïkis of vascular adherent cells. Active proteases can either be secreted directly by inflammatory cells, as elastase and cathepsin G by polymorphonuclear leukocytes, chymase and tryptase by mast cells, and granzymes by lymphocytes, or generated from circulating zymogens by activation in close contact with the cells. This is the case for the pericellular conversion of plasminogen to plasmin, which degrades fibronectin and induces anoïkis of smooth muscle cells. Involvement of proteases has also been proposed in the apoptotic response of cultured adherent cells to serum starvation. Anoïkis is probably involved in pathological remodeling of cardiovascular tissues, including cardiac myocyte detachment in heart failure, deendothelialization and plaque rupture in atherosclerosis, and smooth muscle cell disappearance in aneurysms and varicose veins. The absence of cell adhesion and growth resulting from cleavage of adhesive proteins also represents a major impediment to cellular healing, including the absence of cell recolonization of proteolytically injured tissue and the low efficacy of cell transplantation. However, the exact role of anoïkis in cardiovascular pathologies remains to be further defined.
Collapse
Affiliation(s)
- Jean-Baptiste Michel
- INSERM Unit 460, CHU Xavier Bichat, 46, rue Henri Huchard, 75877 Paris Cedex 18, France.
| |
Collapse
|
34
|
Leidenfrost JE, Khan MF, Boc KP, Villa BR, Collins ET, Parks WC, Abendschein DR, Choi ET. A model of primary atherosclerosis and post-angioplasty restenosis in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:773-8. [PMID: 12875996 PMCID: PMC1868197 DOI: 10.1016/s0002-9440(10)63704-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Although mice deficient in various genes are providing greater insight into the mechanisms of restenosis after angioplasty, there have been limitations with murine models not simulating human vascular disease. To develop a more clinically applicable model of primary atherosclerosis and restenosis following angioplasty of the primary lesion, we fed apolipoprotein E-deficient mice a Western diet and occluded the left common carotid artery for 2 days. Three weeks after flow was restored, the temporarily occluded carotids demonstrated atherosclerotic lesions containing foam cells, cholesterol clefts, necrotic cores, and fibrous capsules. The atherosclerotic carotids in other animals underwent angioplasty with a beaded probe, resulting in plaque and medial layer disruption. Three weeks after angioplasty, although there was significant neointimal lesion formation, the luminal narrowing did not change significantly secondary to overall vessel enlargement (positive remodeling). Neointimal lesions were composed of smooth-muscle cells and extracellular matrix observed adjacent to the original atherosclerotic plaques. Similarly, even at 3 months after the angioplasty the lumen was maintained despite greater neointimal lesion formation caused by progressive positive remodeling. This new murine model of primary atherosclerosis and postangioplasty intimal hyperplasia and remodeling mimics the human disease pattern of postangioplasty intimal hyperplasia. Used in transgenic animals, this model will likely facilitate understanding of the mechanisms of restenosis in humans.
Collapse
Affiliation(s)
- Jeremy E Leidenfrost
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Point - Counterpoint: The wide use of IIb/IIIa inhibitors in interventional cardiology - is it justified? INTERNATIONAL JOURNAL OF CARDIOVASCULAR INTERVENTIONS 2003; 2:61-70. [PMID: 12623390 DOI: 10.1080/acc.2.1.61.70] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
36
|
Kanzaki T, Otabe M. Latent transforming growth factor-beta binding protein-1, a component of latent transforming growth factor-beta complex, accelerates the migration of aortic smooth muscle cells in diabetic rats through integrin-beta3. Diabetes 2003; 52:824-8. [PMID: 12606526 DOI: 10.2337/diabetes.52.3.824] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Aortic smooth muscle cells (SMCs) of diabetic animals have unique properties, including the overexpression of transforming growth factor-beta (TGF-beta) type II receptor, fibronectin, and platelet-derived growth factor beta-receptor. TGF-beta1 is produced and secreted as latent high-molecular weight complex consisting of mature TGF-beta1, latency-associated peptide (LAP), and a latent TGF-beta1 binding protein (LTBP-1). LAP has an important function in the latency of TGF-beta complex, but the role of LTBP-1 is not known in diabetic angiopathy. SMC migration from the medial layer to the intimal layer of an artery is an initial major process of the formation of intimal thickening of an artery. Migration activities of SMCs from diabetic rat with 1-500 pg/ml of LTBP-1 increased significantly compared with that without LTBP-1. LTBP-1 at 10-500 pg/ml stimulated the migration of diabetic SMCs more than SMCs from control rat. An anti-integrin-beta(3) antibody reduced LTBP-1-stimulated migration of diabetic SMCs to 51% compared with no antibody, but it did not reduce that of control SMCs. Furthermore, cross-linking experiments show that LTBP-1 binds integrin-beta(3) in diabetic SMCs much more than in control SMCs in coincidence with the increase of integrin-beta(3) in diabetic aorta by immunohistochemistry. Taken together, these observations suggest that LTBP-1 plays a critical role in intimal thickening of diabetic artery through the acceleration of SMC migration via integrin-beta(3).
Collapse
Affiliation(s)
- Tetsuto Kanzaki
- Department of Internal Medicine, Kohnodai Hospital, National Center of Neurology and Psychiatry, 1-7-1 Kohnodai, Ichikawa City 272-8516, Japan.
| | | |
Collapse
|
37
|
Stawowy P, Graf K, Goetze S, Roser M, Chrétien M, Seidah NG, Fleck E, Marcinkiewicz M. Coordinated regulation and colocalization of alphav integrin and its activating enzyme proprotein convertase PC5 in vivo. Histochem Cell Biol 2003; 119:239-45. [PMID: 12649739 DOI: 10.1007/s00418-003-0506-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2003] [Indexed: 10/25/2022]
Abstract
Integrin alphav is involved in intracellular-extracellular signaling important for cytoskeleton alterations and control of cell movement. In vitro experiments indicate that the integrin alphav-subunit undergoes post-translational endoproteolytic cleavage. This type of activation requires the presence of suitable kexin/subtilisin-like proprotein convertases. In vitro experiments have demonstrated that, among several proprotein convertases, PC5A, and to a threefold lesser extent furin, can activate alphav integrin. The biological significance of these in vitro data would be further supported by a coexpression and coordinated regulation of the gene expression of alphav integrin and its activating enzyme PC5 in vivo. In the present study we investigated the regulation of alphav integrin and PC5 following balloon injury in vivo. Comparative immunocytochemistry revealed a coordinated regulation of alphav integrin and PC5 during vascular remodeling in rodents. Integrin alphav was found to be upregulated in PCNA-positive, proliferating vascular smooth muscle cells. Northern blots revealed no significant regulation of furin mRNA, whereas PC5A mRNA increased during vascular remodeling, suggesting that PC5 is the major convertase during neointima formation in vivo. Incubation of vascular smooth muscle cells with the Golgi-disturbing agent brefeldin A inhibited alphav integrin maturation, indicating that endoproteolytic cleavage occurs in the trans-Golgi network, were PC5 is localized. Thus, the present study further supports the concept that activation of alphav integrin occurs in the trans-Golgi network in vascular smooth muscle cells and involves PC5.
Collapse
Affiliation(s)
- Philipp Stawowy
- Department of Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Various integrin antagonist candidates including antibodies, cyclic peptides, peptidomimetics, and non-peptides have been clinically evaluated and shown to successfully modulate certain disease processes. This review will focus on the key role of the alphav integrin (alphavbeta3 and alphavbeta5) in vascular disorders such as restenosis and angiogenesis-mediated disorders.
Collapse
Affiliation(s)
- Shaker A Mousa
- Albany College of Pharmacy, Albany, New York 12208-3942, USA.
| |
Collapse
|
39
|
Affiliation(s)
- W Cwikiel
- Department of Radiology, University of Michigan Hospital, Ann Arbor 48109, USA
| |
Collapse
|
40
|
Margolin L, Fishbein I, Banai S, Golomb G, Reich R, Perez LS, Gertz SD. Metalloproteinase inhibitor attenuates neointima formation and constrictive remodeling after angioplasty in rats: augmentative effect of alpha(v)beta(3) receptor blockade. Atherosclerosis 2002; 163:269-77. [PMID: 12052473 DOI: 10.1016/s0021-9150(02)00035-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Release of matrix metalloproteinases (MMP) from smooth muscle and foam cells following arterial injury facilitates cell migration, neointimal hyperplasia, and vessel wall remodeling. Inhibition of MMP activity using the hydroxamate, zinc-chelating mimicers of collagen, Batimastat and Marimastat, has shown efficacy in reducing constrictive vascular remodeling 6 weeks after experimental angioplasty but not intimal hyperplasia. Vitronectin receptor (alpha(v)beta(3)) blockade interferes with binding of this integrin to MMP-2 and proteolyzed collagen, thereby reducing cell invasion. This study tests the effect of MMP inhibition, with and without vitronectin receptor (alpha(v)beta(3)) blockade, on neointima formation and arterial remodeling in a long-term model (up to 212 months) of balloon injury in vivo. Male Sabra rats were treated with Batimastat (BB-94, British Biotech Pharmaceuticals Ltd., 30 mg/kg, intraperitoneally) and/or the alpha(v)beta(3) receptor inhibiting RGD peptide, G-Pen-GRGDSPCA (GIBCO BRL, 0.1 micromol), administered as a perivascular gel to the common carotid artery after balloon injury. Animals were sacrificed 3, 14, 25, and 75 days (n=21, 23, 22, and 21) after injury. Animals treated with BB-94, peptide, or both had markedly increased absolute luminal area with markedly reduced luminal cross-sectional-area narrowing by neointima and intima-to-media area ratio at all time points except for 3 days after balloon injury versus non-treated, ballooned animals. Combined treatment was significantly more effective than either one alone. Constrictive remodeling, most marked 212 months after balloon injury, was prevented at this time point in all treated animals. The pattern of reduction in luminal narrowing, neointimal formation, and constrictive remodeling across treatment groups correlated very significantly with the reduction in tissue MMP activity as determined by zymography at 3 days. Confirmation of the efficacy of this strategy in larger animals should be the next step toward testing the applicability of this novel approach to the interventional setting.
Collapse
Affiliation(s)
- Leon Margolin
- Department of Anatomy and Cell Biology, The Hebrew University, Hadassah Medical School, POB 12272, Jerusalem 91120, Israel
| | | | | | | | | | | | | |
Collapse
|
41
|
Vijayakumar E. New drugs: fenoldopam mesylate, glycoprotein IIb/IIIa antagonists, and K+ATP-channel agonists. Int Anesthesiol Clin 2002; 40:35-60. [PMID: 11910249 DOI: 10.1097/00004311-200201000-00005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Kronenwett R, Gräf T, Nedbal W, Weber M, Steidl U, Rohr UP, Möhler T, Haas R. Inhibition of angiogenesis in vitro by alphav integrin-directed antisense oligonucleotides. Cancer Gene Ther 2002; 9:587-96. [PMID: 12082459 DOI: 10.1038/sj.cgt.7700474] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2002] [Indexed: 11/09/2022]
Abstract
The integrin alpha v beta 3 plays a central role in angiogenesis. In this study, we used antisense oligodeoxyribonucleotides (ONs) directed against the alpha v subunit of alpha v beta 3 to inhibit integrin expression. Ten ON sequences, which were selected by systematic alignment of computer-predicted secondary structures of alpha v mRNA, were transfected into human umbilical vein endothelial cells (HUVECs). Following stimulation by PMA, five antisense ONs significantly inhibited alpha v mRNA and protein expression in activated HUVEC at a concentration of 0.05 mciroM with complete prevention of PMA-induced alpha v up-regulation by the most potent antisense ON. Inhibition of alpha v expression was associated with significant inhibition of migration of HUVEC by 28% and had no effect on proliferation and apoptosis. Moreover, transfection of antisense ON inhibited the formation of tube-like structures of HUVEC in Matrigel by 44%. In a cell culture model of angiogenesis consisting of a co-culture of endothelial cells with fibroblasts, transfection of antisense ONs resulted in an inhibition of tube formation of 61%. In conclusion, alpha v antisense ONs are potent inhibitors of angiogenesis in vitro. They might, therefore, be a therapeutic alternative to antagonists, which directly bind to alpha v integrins, and might be useful for the treatment of malignant tumors and hematological malignancies.
Collapse
Affiliation(s)
- Ralf Kronenwett
- Department of Hematology, Oncology and Clinical Immunology, University of Düsseldorf, Düsseldorf, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kenagy RD, Vesti BR, Clowes AW. The urokinase receptor mediates basic fibroblast growth factor-dependent smooth muscle cell migration through baboon aortic explants. Atherosclerosis 2002; 162:63-7. [PMID: 11947898 DOI: 10.1016/s0021-9150(01)00682-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The urokinase receptor is required for vascular smooth muscle cell migration in vitro, but may not be needed in vivo since smooth muscle cell migration and intimal hyperplasia after arterial injury in mice are not affected by urokinase receptor gene deletion. We have used baboon aortic explants as a bridge between cell culture and in vivo experiments to determine if the urokinase receptor is required for smooth muscle cell proliferation and smooth muscle cell migration in primate vessels. Levels of urokinase receptor in explants increased with time after explantation, while blockade of urokinase receptor with an antibody decreased smooth muscle cell proliferation and smooth muscle cell migration from the explants. A blocking antibody to basic fibroblast growth decreased levels of urokinase and urokinase receptor in explants, and it decreased smooth muscle cell migration and mitogenesis. These results suggest that the factor urokinase receptor plays a positive role in smooth muscle cell migration and proliferation in injured primate arterial tissue, in part mediating the pro-migratory and proliferative effects of basic fibroblast growth factor released by damaged smooth muscle cells.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Aorta, Thoracic/cytology
- Aorta, Thoracic/metabolism
- Cell Movement/drug effects
- Dose-Response Relationship, Immunologic
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Enzyme Activation/drug effects
- Fibroblast Growth Factor 2/drug effects
- Fibroblast Growth Factor 2/physiology
- Matrix Metalloproteinase 2/drug effects
- Models, Animal
- Models, Cardiovascular
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Papio
- Plasminogen Activators/drug effects
- Plasminogen Activators/physiology
- Receptors, Cell Surface/drug effects
- Receptors, Cell Surface/physiology
- Receptors, Urokinase Plasminogen Activator
- Urokinase-Type Plasminogen Activator/drug effects
- Urokinase-Type Plasminogen Activator/physiology
Collapse
Affiliation(s)
- R D Kenagy
- Department of Surgery, School of Medicine, University of Washington, Box 356410, AA404, Health Sciences Building, Seattle, WA 98195-6410, USA.
| | | | | |
Collapse
|
44
|
Hong SY, Koh YS, Chung KH, Kim DS. Snake venom disintegrin, saxatilin, inhibits platelet aggregation, human umbilical vein endothelial cell proliferation, and smooth muscle cell migration. Thromb Res 2002; 105:79-86. [PMID: 11864711 DOI: 10.1016/s0049-3848(01)00416-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A novel disintegrin, saxatilin, was purified from Korean snake (Gloydius saxatilis) venom by means of chromatographic fractionations. We have also isolated the cDNA encoding the disintegrin using cDNA library of the snake venom gland and analyzed its complete nucleotide sequence. Saxatilin is a single-chain polypeptide composed of 73 amino acids including 12 cysteines as well as the tripeptide sequence Arg-Gly-Asp (RGD), a proposed recognition site of adhesive proteins. Molecular mass of saxatilin was determined to be 7712 Da by matrix-assisted laser desorption ionization mass spectrometry. Saxatilin inhibits glycoprotein (GP) IIb-IIIa binding to immobilized fibrinogen with IC(50) of 2.0 nM and ADP-induced platelet aggregation with IC(50) of 127 nM, respectively. The snake venom disintegrin also significantly suppresses basic fibroblast growth factor-induced human umbilical vein endothelial cell (HUVEC) proliferation, but has little effect on normal growth of the cell. Interaction of human umbilical vein cell to immobilized vitronectin is also inhibited by binding of saxatilin to alpha(v)beta(3) integrin. Adhesion of smooth muscle cells (SMCs) to vitronectin as well as vitronectin-induced migration of the cells was strongly inhibited by saxatilin. Several lines of experimental evidence suggest potential use of saxatilin for development of therapeutic agents.
Collapse
Affiliation(s)
- Sung-Yu Hong
- Department of Biochemistry, College of Science, and Bioproducts Research Center, Yonsei University, Seoul 120-749, South Korea
| | | | | | | |
Collapse
|
45
|
Fishbein I, Brauner R, Chorny M, Gao J, Chen X, Laks H, Golomb G. Local delivery of mithramycin restores vascular reactivity and inhibits neointimal formation in injured arteries and vascular grafts. J Control Release 2001; 77:167-81. [PMID: 11733085 DOI: 10.1016/s0168-3659(01)00472-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Arterial restenosis is responsible for the high failure rates of vascular reconstruction procedures. Local sustained drug delivery has shown promise in the prevention of restenosis. The drug release rate from mithramycin-loaded EVA matrices (0.1%) was evaluated, and their antirestenotic effect was studied in the rat carotid model and rabbit model of vascular grafts. The modulation of c-myc expression by mithramycin treatment was examined by immunohistochemistry in the rat carotid model. The proliferative response of injured rat arteries was studied by bromdeoxyuridine (BrdU) immunostaining. The impact of mithramycin treatment on vasomotor responses of the venous segments grafted into arterial circulation was studied ex vivo using vasoreactive compounds. Mithramycin was released exponentially from EVA matrices in PBS. Matrices co-formulated with PEG-4600 revealed enhanced release kinetics. The perivascular implantation of drug-loaded EVA-PEG matrices led to 50% reduction of neointimal formation, and reduced the c-myc expression and BrdU labeling in comparison to control implants. Decreased sensitivity of mithramycin-treated grafts to serotonin-induced vasoconstriction was observed. Local perivascular mithramycin treatment limits the functional alteration caused by the grafting of venous segments in high-pressure arterial environment, and potently inhibits stenosis secondary to grafting and angioplasty injury. The antirestenotic effect is associated with reduced c-myc expression and with subsequent decrease in SMC proliferation.
Collapse
Affiliation(s)
- I Fishbein
- Department of Pharmaceutics, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, POB 12065, 91120, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
46
|
Lele M, Sajid M, Wajih N, Stouffer GA. Eptifibatide and 7E3, but not tirofiban, inhibit alpha(v)beta(3) integrin-mediated binding of smooth muscle cells to thrombospondin and prothrombin. Circulation 2001; 104:582-7. [PMID: 11479257 DOI: 10.1161/hc3101.092199] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Our objective was to determine whether abciximab, eptifibatide, or tirofiban inhibited ligand binding to alpha(v)beta(3) integrins on human aortic smooth muscle cells (HASMCs) or human umbilical vein endothelial cells (HUVECs). Abciximab binds alpha(IIb)beta(3) on platelets and alpha(v)beta(3) on HUVECs with similar affinity, whereas eptifibatide and tirofiban are thought to be highly specific for alpha(IIb)beta(3). The conclusion that eptifibatide does not bind vascular alpha(v)beta(3) integrins may be premature, however, because recent studies have demonstrated that the affinity of alpha(v)beta(3) for various ligands, including antagonists, is subject to modulation. METHODS AND RESULTS Abciximab and 7E3, the anti-beta(3) integrin monoclonal antibody from which abciximab was derived, bound alpha(v)beta(3) on HASMCs in a specific and saturable manner and with an affinity similar to binding to alpha(IIb)beta(3) on platelets. 7E3 and eptifibatide inhibited alpha(v)beta(3)-mediated attachment of HASMCs to thrombospondin (TSP) and prothrombin but had no effect on alpha(v)beta(5)- or beta(1)-mediated HASMC attachment to vitronectin-, collagen-, or fibronectin-coated or noncoated tissue culture plates. The inhibitory effect of eptifibatide was similar in magnitude and not additive to that of 7E3. Eptifibatide and 7E3 inhibited alpha(v)beta(3)-mediated attachment of HUVECs. Tirofiban had only nonspecific effects on HASMC attachment to extracellular matrix proteins. In cell proliferation assays, eptifibatide inhibited alpha(v)beta(3)-mediated responses to soluble TSP by HASMCs and beta(3) integrin-expressing HEK cells. CONCLUSIONS Eptifibatide and 7E3, but not tirofiban, specifically inhibit alpha(v)beta(3)-mediated binding of human smooth muscle and endothelial cells.
Collapse
MESH Headings
- Abciximab
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Binding, Competitive/drug effects
- Cell Adhesion/drug effects
- Cell Division/drug effects
- Cell Line
- Cells, Cultured
- Dimerization
- Dose-Response Relationship, Drug
- Eptifibatide
- Humans
- Immunoglobulin Fab Fragments/metabolism
- Immunoglobulin Fab Fragments/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Peptides/metabolism
- Peptides/pharmacology
- Platelet Aggregation Inhibitors/metabolism
- Platelet Aggregation Inhibitors/pharmacology
- Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors
- Prothrombin/metabolism
- Receptors, Vitronectin/antagonists & inhibitors
- Receptors, Vitronectin/chemistry
- Receptors, Vitronectin/physiology
- Thrombospondin 1/metabolism
- Thrombospondin 1/pharmacology
- Thrombospondins/metabolism
- Thrombospondins/pharmacology
- Tirofiban
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
- Tyrosine/pharmacology
- Vitronectin/metabolism
- Vitronectin/pharmacology
Collapse
Affiliation(s)
- M Lele
- Program in Molecular Cardiology, University of North Carolina, Chapel Hill, NC 27599-7075, USA
| | | | | | | |
Collapse
|
47
|
Liu G, Eskin SG, Mikos AG. Integrin alpha(v)beta(3) is involved in stimulated migration of vascular adventitial fibroblasts by basic fibroblast growth factor but not platelet-derived growth factor. J Cell Biochem 2001; 83:129-35. [PMID: 11500961 DOI: 10.1002/jcb.1208] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We examined the effects of basic fibroblast growth factor (bFGF) and platelet-derived growth factor (PDGF) on the migration of vascular adventitial fibroblasts (VAFs) isolated from rat aortic adventitiae. Both bFGF and PDGF significantly stimulated VAF migration in vitro. An antibody to rat beta(3) integrin reduced bFGF-stimulated migration in a dose dependent manner. Moreover, VAF migration was inhibited in the presence of cyclic RGD (cRGD) peptide. However, PDGF-directed migration was blocked only by equivalent cRGD peptide but not by antibody to beta(3) integrin. These data suggest that alpha(v)beta(3) integrin mediates VAF migration stimulated by bFGF and that chemoattractant directed migration may be through distinct integrins.
Collapse
Affiliation(s)
- G Liu
- Department of Bioengineering, Rice University, Houston, TX 77251-1892, USA
| | | | | |
Collapse
|
48
|
Bishop GG, McPherson JA, Sanders JM, Hesselbacher SE, Feldman MJ, McNamara CA, Gimple LW, Powers ER, Mousa SA, Sarembock IJ. Selective alpha(v)beta(3)-receptor blockade reduces macrophage infiltration and restenosis after balloon angioplasty in the atherosclerotic rabbit. Circulation 2001; 103:1906-11. [PMID: 11294811 DOI: 10.1161/01.cir.103.14.1906] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND alpha(v)beta(3)-Integrin receptors are upregulated in atherosclerotic arteries and play a key role in smooth muscle cell and possibly inflammatory cell migration. We hypothesized that after balloon angioplasty (BA) of atherosclerotic arteries, selective inhibition of the alpha(v)beta(3)-receptor by XT199, a small-molecule, non-peptide-selective alpha(v)beta(3)-receptor antagonist, would reduce restenosis. METHODS AND RESULTS After induction of focal atherosclerosis, rabbits underwent femoral BA and received XT199 (2.5 mg/kg IV bolus plus 2.5 mg. kg(-1). d(-1) IV; n=19) or vehicle (n=20) for 14 days. At 28 days after BA, the XT199 group had a larger lumen (0.75+/-0.26 versus 0.57+/-0.20 mm(2), P=0.03) and a smaller neointimal area (0.49+/-0.18 versus 0.68+/-0.25 mm(2), P=0.01) than the vehicle group. Angiographic analysis confirmed a 30% to 40% reduction in restenosis. Arteries harvested at 28 days after BA did not show a reduction in intima plus media smooth muscle cell content but did show a 50% reduction in macrophage cell density in the XT199 group (716+/-452 versus 1458+/-989 cells/mm(2), P<0.006). Neovessel density at 28 days was also reduced (23+/-42 versus 58+/-46 vessel cross sections/mm(2), P<0.02). Early after BA (ie, 3 to 7 days), there was a decrease in intracellular adhesion molecule-1 and vascular cell adhesion molecule-1 expression, indicative of a reduction in vascular cell activation. CONCLUSIONS Selective alpha(v)beta(3)-receptor blockade for 14 days after BA in the focally atherosclerotic rabbit significantly reduced restenosis and limited macrophage infiltration and neovascularization in the vessel wall.
Collapse
MESH Headings
- Actins/analysis
- Angioplasty, Balloon
- Animals
- Arterial Occlusive Diseases/pathology
- Arterial Occlusive Diseases/prevention & control
- Arteriosclerosis/pathology
- Arteriosclerosis/therapy
- Cell Adhesion/drug effects
- Cells, Cultured
- Chemokine CCL2/metabolism
- Cholesterol/blood
- Dose-Response Relationship, Drug
- Femoral Artery/drug effects
- Femoral Artery/metabolism
- Femoral Artery/pathology
- Imidazoles/pharmacology
- Immunohistochemistry
- Intercellular Adhesion Molecule-1/drug effects
- Intercellular Adhesion Molecule-1/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Rabbits
- Receptors, Vitronectin/antagonists & inhibitors
- Receptors, Vitronectin/metabolism
- Recurrence
- Time Factors
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
- Tunica Media/drug effects
- Tunica Media/metabolism
- Tunica Media/pathology
- Vascular Cell Adhesion Molecule-1/drug effects
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- G G Bishop
- University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sheu JR, Wu CH, Chen YC, Hsiao G, Lin CH. Mechanisms in the inhibition of neointimal hyperplasia with triflavin in a rat model of balloon angioplasty. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2001; 137:270-8. [PMID: 11283521 DOI: 10.1067/mlc.2001.114065] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
RGD-containing peptides are able to inhibit the binding of ligands to certain beta3 integrins, such as alpha(IIb)beta3 and alpha(v)beta3, both of which are involved in neointimal hyperplasia. The present study was designed to elucidate the detailed mechanisms involved in the inhibition of neointimal hyperplasia with triflavin in a rat model of balloon angioplasty. Triflavin (0.25 mg x kg(-1) x d(-1)), an RGD-containing disintegrin, time dependently inhibited both neointimal hyperplasia and lumen occlusion after angioplasty in carotid arteries of rats. Furthermore, electron micrographs highlighted that SMCs were phenotypically different from the typical contractile, spindle-shaped SMCs normally seen in uninjured vessel walls. PDGF-BB was strongly produced in thrombus formation and neointimal SMCs after angioplasty, and triflavin significantly reduced PDGF-BB expression in vessel lumens and neointimal SMCs after angioplasty. Balloon angioplasty caused a significant increase of nitrate and cyclic guanosine monophosphate levels compared with levels found in sham-operated rats, and these were not significantly changed with infusion of triflavin (0.25 mg x kg(-1) x d(-1)). Furthermore, the plasma level of TXB2 obviously increased after angioplasty, and triflavin markedly suppressed the elevation of plasma TXB2 concentration. The results indicate that triflavin effectively prevents neointimal hyperplasia, possibly through the following 2 mechanisms. First, triflavin binds to alpha(IIb)beta3 integrin on platelet membranes, resulting in inhibition of platelet adhesion, secretion, and aggregation in injured arteries, followed by inhibition of TXA2 formation and PDGF-BB release from platelets. Second, triflavin may also bind to alpha(v)beta3 integrin on SMCs, thus subsequently inhibiting cell migration and proliferation. These results provide new insights into the mechanisms of neointimal hyperplasia and have significant implications for disintegrin therapy for the treatment of restenosis and atherosclerosis.
Collapse
Affiliation(s)
- J R Sheu
- Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
50
|
Degryse B, Orlando S, Resnati M, Rabbani SA, Blasi F. Urokinase/urokinase receptor and vitronectin/αvβ3 integrin induce chemotaxis and cytoskeleton reorganization through different signaling pathways. Oncogene 2001; 20:2032-43. [PMID: 11360187 DOI: 10.1038/sj.onc.1204261] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2000] [Revised: 01/05/2001] [Accepted: 01/15/2001] [Indexed: 11/09/2022]
Abstract
Vitronectin (VN) and pro-urokinase (pro-uPA) stimulated migration of rat smooth muscle cells in a dose-dependent and additive way, and induced motile-type changes in cell morphology together with a complete reorganization of the actin filaments and of the microtubules. All these effects were inhibited by pertussis toxin, or by antibodies directed against the urokinase receptor (uPAR) or against the VN receptor alpha(v)beta(3) suggesting that an association between the two receptors is required to mediate both signals. Investigation of the signaling pathways showed that increasing the intracellular cAMP resulted in a selective inhibition of VN-induced cell migration. On the other hand, PD 98059, an inhibitor of MEK, differentially inhibited the pro-uPA- but not the VN-induced cell migration. Phosphorylation and nuclear translocation of Erk by pro-uPA was directly observed. We conclude that the signaling pathways of pro-uPA and VN must be at least in part different.
Collapse
Affiliation(s)
- B Degryse
- Department of Molecular Pathology and Medicine, Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | | | | | | | | |
Collapse
|