Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Korean Med Sci ; 36(40): e259, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34664799

ABSTRACT

BACKGROUND: Titanium dioxide films exhibit good biocompatibility and may be effective as drug-binding matrices for drug-eluting stents. We conducted a mid-term evaluation of a novel polymer-free everolimus-eluting stent using nitrogen-doped titanium dioxide film deposition (TIGEREVOLUTION®) in comparison with a commercial durable polymer everolimus-eluting stent (XIENCE Alpine®) in a porcine coronary restenosis model. METHODS: Twenty-eight coronary arteries from 14 mini-pigs were randomly allocated to TIGEREVOLUTION® stent and XIENCE Alpine® stent groups. The stents were implanted in the coronary artery at a 1.1-1.2:1 stent-to-artery ratio. Eleven stented coronary arteries in each group were finally analyzed using coronary angiography, optical coherence tomography, and histopathologic evaluation 6 months after stenting. RESULTS: Quantitative coronary analysis showed no significant differences in the pre-procedural, post-procedural, and 6-month lumen diameters between the groups. In the volumetric analysis of optical coherence tomography at 6 months, no significant differences were observed in stent volume, lumen volume, and percent area stenosis between the groups. There were no significant differences in injury score, inflammation score, or fibrin score between the groups, although the fibrin score was zero in the TIGEREVOLUTION® stent group (0 vs. 0.07 ± 0.11, P = 0.180). CONCLUSION: Preclinical evaluation, including optical coherence tomographic findings 6 months after stenting, demonstrated that the TIGEREVOLUTION® stent exhibited efficacy and safety comparable with the XIENCE Alpine® stent, supporting the need for further clinical studies on the TIGEREVOLUTION® stent.


Subject(s)
Coronary Restenosis/drug therapy , Drug-Eluting Stents , Everolimus/therapeutic use , Animals , Coronary Angiography , Coronary Restenosis/pathology , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Everolimus/chemistry , Polymers/chemistry , Swine , Swine, Miniature , Titanium/chemistry , Tomography, Optical Coherence
2.
Article in English | MEDLINE | ID: mdl-30453089

ABSTRACT

Restenosis is a major problem after percutaneous coronary intervention (PCI) treatment. Inflammation is one of the major core mechanisms involved in the occurrence of restenosis, and plays an important role in intimal hyperplasia. Detoxification and activating blood circulation decoction (DABCD) is a traditional Chinese medicine that is used in the treatment and prevention of atherosclerotic and inflammatory diseases. Our previous studies demonstrated that DABCD-mediated cardioprotection involves anti-inflammatory mechanisms and could be developed as a novel drug for the treatment of vascular smooth muscle cell (VSMC) proliferation and aortic restenosis. A rat model of postoperative restenosis after PCI was generated by balloon injury to determine the protective effects and potential mechanisms of DABCD. The injured segments of aortae were collected on days 14 and 28 after the operation to observe the morphological changes in the vascular structure and measure the proportion of inflammatory factors in plasma and vascular tissues, as well as test the proliferative activity of VSMCs. The expression of related proteins, namely, Toll-like receptor (TLR) 4 and nuclear factor (NF)-κB, in the mechanistic study was clarified by western blot analysis. We tested the hypothesis that the cardioprotective effects of DABCD on aortic restenosis are associated with the inhibition of aortic intimal hyperplasia in this model. Our results showed that DABCD has protective effect on rat aortic restenosis and the anti-inflammatory mechanism of DABCD on balloon-induced restenosis in rat may be due to its ability to inhibit TLR4-mediated NF-κB signaling pathways. DABCD may be a potential therapeutic agent against restenosis.


Subject(s)
Balloon Occlusion/adverse effects , Blood Circulation/drug effects , Coronary Restenosis/physiopathology , Drugs, Chinese Herbal/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Cell Proliferation/drug effects , Coronary Restenosis/etiology , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Cytokines/metabolism , Drugs, Chinese Herbal/therapeutic use , Gene Expression Regulation/drug effects , Male , Muscle, Smooth, Vascular/pathology , Rats , Rats, Sprague-Dawley
3.
Sci Rep ; 6: 29975, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27453531

ABSTRACT

Restenosis limits the efficacy of vascular percutaneous intervention, in which vascular smooth muscle cell (VSMC) proliferation and activation of inflammation are two primary causal factors. Calpains influence VSMC proliferation and collagen synthesis. However, the roles of calpastatin and calpains in vascular restenosis remain unclear. Here, restenosis was induced by ligating the left carotid artery, and VSMCs were pretreated with platelet-derived growth factor (PDGF)-BB. Adenovirus vector carrying MMP2 sequence and specific small interfering RNA against calpain-1/2 were introduced. Finally, restenosis enhanced the expression of calpain-1/2, but reduced calpastatin content. In calpastatin transgenic mice, lumen narrowing was attenuated gradually and peaked on days 14-21. Cell proliferation and migration as well as collagen synthesis were inhibited in transgenic mice, and expression of calpain-1/2 and MMP2/transforming growth factor-ß1 (TGF-ß1). Consistently, in VSMCs pretreated with PDGF-BB, calpastatin induction and calpains inhibition suppressed the proliferation and migration of VSMCs and collagen synthesis, and reduced expression of calpain-1/2 and MMP2/TGF-ß1. Moreover, simvastatin improved restenosis indicators by suppressing the HIF-1α/calpains/MMP2/TGF-ß1 pathway. However, MMP2 supplementation eliminated the vascular protection of calpastatin induction and simvastatin. Collectively, calpains inhibition plays crucial roles in vascular restenosis by preventing neointimal hyperplasia at the early stage via suppression of the MMP2/TGF-ß1 pathway.


Subject(s)
Calpain/antagonists & inhibitors , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Matrix Metalloproteinase 2/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Carotid Arteries/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Platelet-Derived Growth Factor/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Simvastatin/pharmacology
4.
Mol Med Rep ; 13(6): 4959-68, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27108914

ABSTRACT

The mechanism and associated factors of restenosis following intravascular stent implantation remain to be elucidated. The present two­part experimental and clinical study aimed to investigate the effects of tripterygium glycosides on in­stent restenosis subsequent to intra­arterial therapy. Following endovascular stent implantation in rabbit iliac arteries, post­stent outcomes were evaluated in cyclosporine groups, low­dose and high­dose tripterygium glycosides groups and controls. Post­operative angiography indicated that vessel diameters were similar between groups; however, at 28 days after receiving the therapeutic agents, vessels of the cyclosporine and tripterygium glycosides groups were significantly larger than those of the controls. Furthermore, three groups of patients had comparable baseline levels of interleukin (IL)­10, IL­18 and C­reactive protein, and intima­media thickness. However, 1 month after stent implantation, levels of IL­10 and IL­18 were markedly reduced in the high­ and low­dose tripterygium glycosides groups compared with controls. At 6 months after surgery, the stent patency rate in patients with bare stents was significantly lower than in patients receiving tripterygium glycosides (P≤0.009). In addition, the ankle­brachial index was also higher than in those without tripterygium glycosides (P<0.001). Results of the experimental and clinical studies suggest that tripterygium glycosides may inhibit and possibly aid in the prevention of in­stent restenosis formation following endovascular treatment of lower­extremity artery disease.


Subject(s)
Endovascular Procedures/adverse effects , Glycosides/therapeutic use , No-Reflow Phenomenon/drug therapy , No-Reflow Phenomenon/etiology , Plant Extracts/therapeutic use , Tripterygium/chemistry , Angiography , Animals , Biomarkers , Case-Control Studies , Constriction, Pathologic/diagnosis , Constriction, Pathologic/drug therapy , Constriction, Pathologic/etiology , Coronary Restenosis/drug therapy , Coronary Restenosis/etiology , Coronary Restenosis/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endovascular Procedures/methods , Gene Expression , Glycosides/administration & dosage , Glycosides/adverse effects , Humans , Iliac Artery/pathology , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Male , No-Reflow Phenomenon/diagnosis , Plant Extracts/administration & dosage , Plant Extracts/adverse effects , Rabbits , Treatment Outcome
5.
Ther Adv Cardiovasc Dis ; 7(4): 214-23, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23813548

ABSTRACT

Acute coronary syndrome has a high mortality rate that dramatically increases in the presence of left main coronary artery (LMCA) disease. Over the past decades, coronary artery bypass graft (CABG) surgery has been commonly accepted as the standard of care for patients with LMCA stenosis and is still considered the first-line treatment in current practice guidelines. Percutaneous coronary intervention (PCI) of protected and unprotected LMCA has gained popularity and is increasingly utilized with comparable outcomes to CABG in randomized controlled trials. In-stent restenosis and the need for revascularization provide the main obstacle to LMCA revascularization. The advent of better PCI equipment, stents, ablative devices, intravascular ultrasound, hemodynamic support devices and antithrombotic agents have ignited a renewed interest in the practice of LMCA PCI, especially for high surgical risk patients who are neither candidates nor agreeable to CABG surgery. Herein, we review the studies comparing unprotected LMCA stenting with CABG surgery in regard to 3 main endpoints: mortality, major adverse events and the incidence of repeat revascularization.


Subject(s)
Acute Coronary Syndrome/therapy , Coronary Artery Bypass/methods , Stents , Acute Coronary Syndrome/surgery , Coronary Restenosis/pathology , Coronary Stenosis/physiopathology , Coronary Stenosis/surgery , Coronary Stenosis/therapy , Equipment Design , Humans , Percutaneous Coronary Intervention/methods , Practice Guidelines as Topic , Risk Factors , Treatment Outcome
6.
Cardiovasc Res ; 93(3): 414-23, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22116621

ABSTRACT

AIMS: Alternative transcription and splicing of the allograft inflammatory factor-1 (AIF-1) gene results in the expression of two different proteins: AIF-1 and interferon responsive transcript-1 (IRT-1).  Here, we explore the impact of AIF-1 and IRT-1 on vascular smooth muscle cell (VSMC) activation and neointima formation, the mechanisms underlying their alternative splicing, and associations of AIF-1 and IRT-1 mRNA with parameters defining human atherosclerotic plaque phenotype. METHODS AND RESULTS: Translation of AIF-1 and IRT-1 results in different products with contrasting cellular distribution and functions. Overexpression of AIF-1 stimulates migration and proliferation of human VSMCs, whereas IRT-1 exerts opposite effects. Adenoviral infection of angioplasty-injured rat carotid arteries with AdAIF-1 exacerbates intima hyperplasia, whereas infection with AdIRT-1 reduces neointima. Expression of these variants is modulated by changes in nuclear factor of activated T-cells (NFAT) activity.  Pharmacological inhibition of NFAT or targeting of NFATc3 with small interfering RNA (siRNA) lowers the AIF-1/IRT-1 ratio and favours an anti-proliferative outcome.  NFAT acts as a repressor on the IRT-1 transcriptional start site, which is also sensitive to interferon-γ stimulation. Expression of AIF-1 mRNA in human carotid plaques associates with less extracellular matrix and a more pro-inflammatory plaque and plasma profile, features that may predispose to plaque rupture. In contrast, expression of IRT-1 mRNA associates with a less aggressive phenotype and less VSMCs at the most stenotic region of the plaque. CONCLUSION: Inhibition of NFAT signalling, by shifting the AIF-1/IRT-1 ratio, may be an attractive target to regulate the VSMC response to injury and manipulate plaque stability in atherosclerosis.


Subject(s)
Alternative Splicing/physiology , Carotid Artery Diseases , Coronary Artery Disease , DNA-Binding Proteins/genetics , NFATC Transcription Factors/metabolism , Neointima , Angioplasty, Balloon, Coronary/adverse effects , Animals , Calcium-Binding Proteins , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Coronary Vessels/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Microfilament Proteins , Muscle, Smooth, Vascular/pathology , Myometrium/blood supply , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
7.
Cell Biochem Funct ; 29(3): 207-14, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21374644

ABSTRACT

Drug-eluting stents (DES) were developed to combat the problem of in-stent restenosis, and evaluating the biological activity from DES systems is critical for its safety and efficacy. To test the cytotoxicity of nitric oxide (NO) donor-containing polymers for their potential use in DES applications, S-nitrosoglutathione (GSNO) or in combination with poly(vinyl alcohol) (PVA) and poly(vinyl pyrrolidone) (PVP) in an aqueous polymeric solution (PVA/PVP/GSNO) was investigated using Balb/c 3T3 and Rabbit arterial smooth muscle (RASM) cells. The sensitivity of 3T3 cells to the cytotoxicity effects induced by GSNO was higher than that of RASM cells, while RASM cells were more susceptible to alterations in membrane permeability. Cell growth assays showed that GSNO and PVA/PVP/GSNO induced antiproliferative effects in RASM cells. Moreover, the presence of polymers can reduce the cytotoxicity and enhance the antiproliferative effects of GSNO. Dose-dependent inhibition of platelet aggregation was similar for both PVA/PVP/GSNO (EC50 of 3.4 ± 2.3 µM) and GSNO (EC50 of 2.8 ± 1.1 µM) solutions. Platelet adhesion assays showed that the inhibition caused by GSNO (EC50 of 5.0 mM) was dependent on the presence of plasma. These results demonstrate that the methodology adopted here is suitable to establish safety margins and evaluate the antithrombotic potential and antiproliferative effects of NO-eluting biomaterials and polymeric solutions for the new cardiovascular devices, and also to emphasize the importance of using more specific cell lines in these evaluations.


Subject(s)
Drug-Eluting Stents , Fibrinolytic Agents/pharmacology , Nitric Oxide Donors/pharmacology , S-Nitrosoglutathione/pharmacology , Animals , BALB 3T3 Cells , Cell Proliferation/drug effects , Coronary Restenosis/drug therapy , Coronary Restenosis/pathology , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions/metabolism , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/therapeutic use , Humans , Mice , Myocytes, Smooth Muscle , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Nitric Oxide Donors/therapeutic use , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Polyvinyl Alcohol/pharmacology , Povidone/pharmacology , Rabbits , S-Nitrosoglutathione/metabolism , S-Nitrosoglutathione/therapeutic use
8.
Am Heart J ; 160(5): 804-11, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21095265

ABSTRACT

BACKGROUND: The optimal duration of clopidogrel therapy after coronary stenting is debated because of the scarcity of randomized controlled trials and inconsistencies arising from registry data. Although prolonged clopidogrel therapy after bare metal stenting is regarded as an effective secondary prevention measure, the safety profile of drug-eluting stents itself has been questioned in patients not receiving ≥ 12 months of dual-antiplatelet therapy. HYPOTHESIS: Twenty-four months of clopidogrel therapy after coronary stenting reduces the composite of death, myocardial infarction, or stroke compared with 6 months of treatment. STUDY DESIGN: PRODIGY is an unblinded, multicenter, 4-by-2 randomized trial. All-comer patients with indication to coronary stenting are randomly treated-balancing randomization-with bare metal stent (no active late loss inhibition), Endeavor Sprint zotarolimus-eluting stent (Medtronic, Santa Rosa, CA) (mild late loss inhibition), Taxus paclitaxel-eluting stent (Boston Scientific, Natick, MA) (moderate late loss inhibition), or Xience V everolimus-eluting stent (Abbott Vascular, Santa Clara, CA) (high late loss inhibition). At 30 days, patients in each stent group are randomly allocated to receive 24 or up to 6 months of clopidogrel therapy-primary end point randomization. With 1,700 individuals, this study will have >80% power to detect a 40% difference in the primary end point after sample size augmentation of 5% and a background event rate of 8%. SUMMARY: The PRODIGY trial aims to assess whether 24 months of clopidogrel therapy improves cardiovascular outcomes after coronary intervention in a broad all-comer patient population receiving a balanced mixture of stents with various anti-intimal hyperplasia potency.


Subject(s)
Angioplasty, Balloon, Coronary/methods , Coronary Disease/surgery , Coronary Restenosis/prevention & control , Coronary Vessels/pathology , Drug-Eluting Stents/adverse effects , Ticlopidine/analogs & derivatives , Tunica Intima/pathology , Clopidogrel , Coronary Disease/drug therapy , Coronary Disease/pathology , Coronary Restenosis/etiology , Coronary Restenosis/pathology , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Follow-Up Studies , Humans , Hyperplasia/etiology , Hyperplasia/pathology , Hyperplasia/prevention & control , Platelet Aggregation Inhibitors/administration & dosage , Ticlopidine/administration & dosage , Time Factors , Treatment Outcome , Tunica Intima/drug effects
9.
Coron Artery Dis ; 21(5): 286-91, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20508518

ABSTRACT

OBJECTIVES: The branched-chain fatty acid, valproic acid (VPA), is the most commonly used anti-epileptic drug for treating generalized epilepsy. Recently antiproliferative effects of VPA have been described in human cancer cells, and phase I trials for the treatment of solid tumors have been initiated. In cardiologic patients, increased cell proliferation and migration from the media into the subendothelial space are the key events causing restenosis after coronary angioplasty and stenting. This study investigates the effect of VPA on proliferation and migration in human coronary vascular cells. METHODS AND RESULTS: The theoretical clinical relevance of the data is estimated with a SI/MPL-ratio, which is defined as the relationship between a significant effect in vitro (SI) and the maximal plasma level in vivo (MPL). Dilution of VPA: Aqua dest, MPL in vivo: 100 microg/ml. Cell culture: HUVEC, human umbilical endothelial cells; HCAEC, human coronary artery endothelial cells; HCMSMC, human coronary media smooth muscle cells. Proliferation assay: HUVEC, HCAEC, and HCMSMC were seeded as described. At day 1, after seeding the cell number was calculated in a cell counter. VPA was added in six different concentrations ranging between 50 and 300 microg/ml. At day 3, the medium and agent were renewed, and after another 2 days, the cell number was calculated in relation with the cell number at day 1. Cell toxicity: Cytotoxic effects of VPA were studied in concentrations ranging from 50 to 300 microg/ml. Migration assay: migration of HCMSMC after incubation with VPA in concentrations ranging from 50 to 300 microg/ml was studied for a period of 24 h. Proliferation assay: strong dose-dependent antiproliferative effects were detected after 5 days of incubation with all the three tested cell types. In HUVEC, significant antiproliferative effects were found with VPA in concentrations of 100 microg/ml (P<0.05, SI/MPL-ratio: 1.0) and more. In HCAEC and HCMSMC, significant antiproliferative effects were detected after incubation with VPA in the concentrations of 50 microg/ml (HCAEC: P<0.01, SI/MPL ratio: 0.5; HCMSMC: P<0.001, SI/MPL-ratio: 0.5). Migration assay: no effect on cell migration was detected after incubation of HCMSMC for a period of 48 h with VPA in concentrations ranging from 50 to 300 microg/ml. Cell toxicity: in HUVEC, HCAEC, and HCMSMC significant toxic effects were detected in all the VPA concentrations studied. CONCLUSION: Significant dose-dependent antiproliferative effects of VPA with SI/MPL ratios of 0.5 identify the drug as a promising candidate for both systemic and local therapy of postinterventional restenosis. The partial cytotoxic effects, however, may restrict the use of VPA to local high-dose devices such as drug eluting stents.


Subject(s)
Cell Proliferation/drug effects , Coronary Restenosis/drug therapy , Endothelial Cells/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Valproic Acid/pharmacology , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Coronary Restenosis/pathology , Coronary Vessels/drug effects , Coronary Vessels/pathology , Dose-Response Relationship, Drug , Endothelial Cells/pathology , Humans , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Time Factors , Valproic Acid/toxicity
10.
Phytomedicine ; 17(3-4): 233-40, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19748258

ABSTRACT

AIM OF THE STUDY: the effect of total saponins of "panax notoginseng root" on aortic intimal hyperplasia and the expressions of cell cycle protein and extracellular matrix in rats MATERIALS AND METHODS: Sprague-Dawley rats were randomly divided into sham-operated, control, TSPN and atorvastatin group. Rat aorta intima in all groups were injured by insertion of domestic balloon catheter into the aortae except sham-operated rats. Drugs were administrated orally from the second day after vascular injury and continued for 14 days. The injured segments of aortae were collected on the sixteenth day after operation to observe the morphological changes of vascular structure and to examine the expressions of proliferating cell nuclear antigen(PCNA), cyclinD1, cyclinE, collagen I(Col-I), fibronect(FN), matrix metalloproteinase-9(MMP-9) and tissue inhibitor metalloproteinase-1(TIMP-1). RESULTS: TPNS significantly inhibited the vascular intimal hyperplasia. TPNS significantly lowered the expression of PCNA, cyclinE, cyclinD1, FN and MMP-9. TPNS had no significant impacts on the expression of Col-I and TIMP-1. CONCLUSIONS: Our studies indicated that TSPN could inhibit vessel restenosis after vascular intimal injury, and its mechanisms may be related to the blockage of the excessive proliferation of VSMC, the reduction of ECM protein deposition in the endometrium, and the degradation of ECM protein.


Subject(s)
Cell Cycle Proteins/metabolism , Coronary Restenosis/prevention & control , Drugs, Chinese Herbal/therapeutic use , Extracellular Matrix/metabolism , Panax notoginseng/chemistry , Saponins/therapeutic use , Tunica Intima/drug effects , Animals , Anticholesteremic Agents , Aorta/drug effects , Aorta/pathology , Atorvastatin , Collagen Type I/metabolism , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Cyclins/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Fibronectins/metabolism , Heptanoic Acids , Hyperplasia/drug therapy , Hyperplasia/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Phytotherapy , Plant Roots , Proliferating Cell Nuclear Antigen/metabolism , Pyrroles , Random Allocation , Rats , Rats, Sprague-Dawley , Saponins/pharmacology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tunica Intima/pathology
11.
Am J Physiol Heart Circ Physiol ; 291(6): H2692-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16731651

ABSTRACT

Stress-induced release of IL-1alpha and fibroblast growth factor-1 is dependent on intracellular copper and is a major driver of neointimal hyperplasia. Therefore, we assessed the effect of tetrathiomolybdate (TTM), a clinically proven copper chelator, on in-stent restenosis. Nine pigs were treated with TTM (5 mg/kg po) twice daily for 2 wk before stent implantation and for 4 wk thereafter, and nine pigs served as controls. In-stent restenosis was assessed by quantitative coronary angiography (QCA), intravascular ultrasound (IVUS), and histomorphometry. Serum ceruloplasmin activity was used as a surrogate marker of copper bioavailability. In TTM-treated animals, ceruloplasmin dropped 70 +/- 10% below baseline levels. Baseline characteristics were comparable in TTM-treated and control animals. At 4-wk follow-up, all parameters relevant to in-stent restenosis were significantly reduced in TTM-treated animals: minimal lumen diameter by QCA was 2.03 +/- 0.57 and 1.47 +/- 0.45 mm in TTM-treated and control animals, respectively (P < 0.05), percent stenosis diameter was 39% less in TTM-treated animals (27.1 +/- 16.6% vs. 44.5 +/- 16.1%, P < 0.05), minimal lumen area by IVUS was 60% larger in TTM-treated animals (4.27 +/- 1.56 vs. 2.67 +/- 1.19 mm(2), P < 0.05), and neointimal volume by histomorphometry was 37% less in TTM-treated animals (34.9 +/- 11.5 vs. 55.2 +/- 19.6 mm(3), P < 0.05). We conclude that systemic copper chelation with a clinically approved chelator significantly inhibits in-stent restenosis.


Subject(s)
Chelating Agents/pharmacology , Copper/metabolism , Coronary Restenosis/prevention & control , Coronary Vessels/physiopathology , Molybdenum/pharmacology , Stents , Animals , Ceruloplasmin/metabolism , Chelating Agents/metabolism , Chelation Therapy/methods , Coronary Angiography , Coronary Restenosis/pathology , Coronary Restenosis/physiopathology , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Disease Models, Animal , Male , Molybdenum/metabolism , Swine , Time Factors , Tunica Intima/drug effects , Tunica Intima/pathology , Ultrasonography, Interventional
12.
Toxicol Pathol ; 34(1): 11-8, 2006.
Article in English | MEDLINE | ID: mdl-16507539

ABSTRACT

Coronary artery disease remains a major problem for Western societies. The advent of percutaneous interventions, including stents has brought clinical care to a new level of efficacy, yet problems remain. Restenosis following stenting in human coronary arteries appears at last to be yielding to therapeutic strategies, especially drug eluting stents. Because therapeutic percutaneous coronary intervention is widely dominated by the intracoronary stent, restenosis therapies must include the stented coronary artery. Animal models and in particular the porcine coronary model seem to represent the human coronary artery reaction to stenting. It mimics several clinical conditions including thrombosis and neointimal formation. A key question in the era of intravascular technologies is how well this and other models can predict clinical events. This paper discusses the models and their application.


Subject(s)
Coronary Artery Disease/pathology , Coronary Restenosis/pathology , Disease Models, Animal , Stents/adverse effects , Animals , Coronary Artery Disease/etiology , Coronary Artery Disease/therapy , Coronary Restenosis/complications , Coronary Restenosis/prevention & control , Coronary Thrombosis/etiology , Coronary Thrombosis/pathology , Coronary Vessels/injuries , Coronary Vessels/pathology , Dogs , Drug Evaluation, Preclinical , Rabbits , Rats , Swine , Tunica Intima/pathology
13.
Article in English | MEDLINE | ID: mdl-15715502

ABSTRACT

Approximately 12 million Americans have coronary artery disease, and almost one in five deaths in the United States can be attributed to this disease. In addition, 1.2 million Americans undergo cardiac catheterization and over one-half million receive a percutaneous coronary intervention such as balloon angioplasty, atherectomy, or stent implantation annually. This article will provide an overview of (1) atherosclerosis, the progressive disease which can lead to thrombotic events and/or the development of hemodynamically significant coronary artery lesions; (2) restenosis, the reappearance of significant lesions after coronary interventions such as stent placement; and (3) drug-eluting stents, the devices which, by using appropriate polymers to elute the appropriate drug with the appropriate pharmacokinetics, have almost completely eliminated restenosis.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Coronary Artery Disease/therapy , Coronary Restenosis/prevention & control , Immunosuppressive Agents/therapeutic use , Stents , Thrombosis/prevention & control , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Clinical Trials as Topic , Coronary Artery Disease/etiology , Coronary Artery Disease/pathology , Coronary Restenosis/etiology , Coronary Restenosis/pathology , Delayed-Action Preparations , Drug Evaluation, Preclinical , Humans , Immunosuppressive Agents/pharmacokinetics , Polymers , Stents/adverse effects , Thrombosis/etiology , Thrombosis/pathology
14.
J Cardiol ; 44(1): 13-20, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15334880

ABSTRACT

BACKGROUND: A single and local administration of L-arginine after balloon angioplasty enhances nitric oxide (NO) generation and inhibits lesion formation in animals. OBJECTIVES: The present study assessed the effect of increasing NO to inhibit restenosis after percutaneous transluminal coronary angioplasty (PTCA) in humans by local and systemic administration of L-arginine, a precursor of NO in humans. METHODS: L-arginine was administered to 34 consecutive patients with angina pectoris or old myocardial infarction via a cardiac catheter (500 mg/4 min) before PTCA, and via a peripheral vein (30 g/4 hr, for 5 days) after PTCA. Patients were treated between December 1998 and December 2000. Plasma concentrations of L-arginine, NO (as nitrite + nitrate) and cyclic guanosine monophosphate (cGMP) were measured before and after L-arginine administration. The control group consisted of 90 patients who underwent PTCA successfully without L-arginine administration in the period between July 1996 and November 1998. Baseline clinical and angiographic characteristics were compared between the two groups. All patients were followed by coronary angiography for 3 months after PTCA. Quantitative coronary angiography and restenosis rate were studied. RESULTS: Baseline clinical and angiographic characteristics were not different between the two study groups. Despite a significant elevation in plasma L-arginine concentration after L-arginine administration, NO and cGMP did not increase significantly. After PTCA, the difference in restenosis rates between L-arginine and control subjects (34% vs 44%) was not significantly different. CONCLUSIONS: Short-term administration of high dose L-arginine did not significantly change the restenosis rate after PTCA.


Subject(s)
Angioplasty, Balloon, Coronary , Arginine/administration & dosage , Coronary Restenosis/drug therapy , Aged , Angina Pectoris/therapy , Coronary Restenosis/blood , Coronary Restenosis/pathology , Cyclic GMP/blood , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Myocardial Infarction/therapy , Nitric Oxide/blood
15.
Arterioscler Thromb Vasc Biol ; 24(8): 1342-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15155381

ABSTRACT

Receptor for AGE (RAGE) is a multi-ligand member of the immunoglobulin superfamily of cell surface molecules. Engagement of RAGE by its signal transduction ligands evokes inflammatory cell infiltration and activation in the vessel wall. In diabetes, when fueled by oxidant stress, hyperglycemia, and superimposed stresses such as hyperlipidemia or acute balloon/endothelial denuding arterial injury, the ligand-RAGE axis amplifies vascular stress and accelerates atherosclerosis and neointimal expansion. In this brief synopsis, we review the use of rodent models to test these concepts. Taken together, our findings support the premise that RAGE is an amplification step in vascular inflammation and acceleration of atherosclerosis. Future studies must rigorously test the potential impact of RAGE blockade in human subjects; such trials are on the horizon.


Subject(s)
Arteriosclerosis/metabolism , Diabetic Angiopathies/metabolism , Glycation End Products, Advanced/metabolism , Models, Animal , Receptors, Immunologic/physiology , Vasculitis/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/pathology , Cell Adhesion Molecules/biosynthesis , Coronary Restenosis/etiology , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Cytokines/biosynthesis , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Angiopathies/pathology , Disease Progression , Drug Evaluation, Preclinical , Hyperlipidemias/blood , Hyperlipidemias/complications , Hyperlipidemias/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Rats , Rats, Zucker , Receptor for Advanced Glycation End Products , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/therapeutic use , Signal Transduction , Solubility , Vasculitis/pathology
16.
Arterioscler Thromb Vasc Biol ; 24(1): 181-6, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14592842

ABSTRACT

OBJECTIVE: We recently demonstrated that Rho-kinase, an effector of the small GTPase Rho, is substantially involved in the pathogenesis of arteriosclerosis. In this study, we examined whether Rho-kinase is also involved in in-stent restenosis and if so, what mechanism is involved. METHODS AND RESULTS: Pigs underwent stent implantation in the left coronary artery with or without administration of fasudil (30 mg/kg per day orally), a specific Rho-kinase inhibitor, starting 2 days before the procedure for a duration of 4 weeks. On day 28, reductions in coronary diameter and neointimal formation associated with macrophage accumulation, collagen deposition, and transforming growth factor (TGF)-beta1 expression were noted at the stent site, and all were significantly suppressed by fasudil. On day 7, fasudil significantly increased the frequency of TUNEL-positive apoptotic cells, while it tended to reduce that of bromodeoxyuridine-positive proliferating cells in the neointima. Western blot analysis on day 7 demonstrated that phosphorylations of the ezrin/radixin/moesin family (a marker of Rho-kinase activity in vivo) and protein expression of monocyte chemoattractant protein-1and bcl-2 were upregulated at the stent site and were significantly suppressed by fasudil. CONCLUSIONS: These results indicate that long-term inhibition of Rho-kinase suppresses in-stent neointimal formation by multiple mechanisms, including reduced vascular inflammation, enhanced apoptosis, and decreased collagen deposition.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Coronary Restenosis/enzymology , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/physiology , Stents , Tunica Intima/pathology , Animals , Apoptosis/drug effects , Blood Proteins/metabolism , Catheterization/adverse effects , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Collagen/metabolism , Coronary Restenosis/pathology , Coronary Restenosis/prevention & control , Coronary Stenosis/surgery , Coronary Stenosis/therapy , Cytoskeletal Proteins/metabolism , Drug Evaluation, Preclinical , Genes, bcl-2 , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Single-Blind Method , Swine , Tunica Intima/drug effects , Tunica Intima/enzymology , rho-Associated Kinases
17.
Cell Mol Biol (Noisy-le-grand) ; 50(8): 953-88, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15704259

ABSTRACT

Several millions of patients with coronary heart disease worldwide are treated by means ofpercutaneous interventions each year. Above all conventional balloon dilation and implantation of uncoated stents are, however, only of limited success as reflected by 6-month restenosis rates of 50% (balloon dilation) and 25-35% (bare-metal stent). It is therefore of utmost importance to identify high-risk groups and explore further secondary-prophylactic measures for the prevention of restenosis. A large body of evidence suggests that elevated homocysteine and/or folate and B-vitamin deficiencies are relevant risk factors for restenoses due to their proatherothrombotic potential. Hyperhomocysteinemia is an ideal target as this parameter can be lowered easily, safely and at a low cost by means of folate and B-vitamin supplementation. The results of published studies exploring a potential correlation between homocysteine levels and the risk of restenosis and those of interventional studies for the reduction of the risk of restenosis have not yet lead to consistent conclusions. However, a critical assessment can by no means exclude the plausibility of postinterventional lowering of homocysteine levels. This review aims at providing insight into the current evidence and biological plausibility of homocysteine-lowering therapy in regard to PCI-related vascular damage. Currently available clinical observational and interventional studies are reviewed in detail.


Subject(s)
Coronary Angiography/methods , Coronary Restenosis/pathology , Homocysteine/physiology , Adult , Aged , Angioplasty , Angioplasty, Balloon, Coronary/methods , Animals , Clinical Trials as Topic , Coronary Artery Disease/pathology , Coronary Stenosis/pathology , Endothelium, Vascular/metabolism , Female , Homocysteine/blood , Homocysteine/metabolism , Homozygote , Humans , Hyperhomocysteinemia/metabolism , Male , Middle Aged , Models, Biological , Risk , Risk Factors , Vitamin B Complex/metabolism
18.
Catheter Cardiovasc Interv ; 57(2): 266-71, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12357534

ABSTRACT

Estrogen can inhibit intimal proliferation and accelerate endothelial regeneration after angioplasty. This suggests that estrogen may prevent in-stent restenosis. Unlike other therapies to prevent restenosis, estrogen may also not delay endothelial regrowth, thereby avoiding the risk of late stent thrombosis. The purpose of this work was to determine the effect of a 17beta-estradiol-eluting stent on neointimal formation in a porcine model. Each artery of six pigs was randomized to either a control, low-dose, or high-dose 17beta-estradiol-eluting stent. All animals were sacrificed at 30 days for histopathological analysis. There was a 40% reduction in intimal area in the high-dose stents compared with control stents (2.54 +/- 1.0 vs. 4.13 +/- 1.1 mm(2), for high dose vs. control, respectively; P < 0.05). There was complete endothelial regeneration at 30 days and similar inflammatory response to stenting on histopathology in all the stent groups. This is the first study to show that 17beta-estradiol-eluting stents are associated with reduced neointimal formation without affecting endothelial regeneration in the pig model of in-stent restenosis. Estrogen-coated stents may have a potential benefit in the prevention and treatment of in-stent restenosis.


Subject(s)
Coated Materials, Biocompatible , Coronary Restenosis/therapy , Drug Delivery Systems , Estradiol/administration & dosage , Stents , Animals , Coronary Restenosis/pathology , Coronary Restenosis/prevention & control , Endothelium, Vascular/pathology , Estradiol/therapeutic use , Models, Animal , Phosphorylcholine , Prosthesis Design , Random Allocation , Swine
19.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 21(8): 591-4, 2001 Aug.
Article in Chinese | MEDLINE | ID: mdl-12575573

ABSTRACT

OBJECTIVE: To study the effect of Xiongshao Capsule (XS) on vascular remodeling in porcine coronary balloon injury model. METHODS: Restenosis model was established by oversized balloon injury at mid-region of the left anterior descending coronary artery. The effect of drugs on late lumen loss and vascular remodeling was evaluated with quantitative histological method by combining results from histopathological analysis and coronary arteriography. RESULTS: Vascular remodeling explained (59 +/- 20)% of late lumen loss. All drugs significantly reduced late lumen loss 4 weeks after balloon injury (P < 0.05 or 0.01). Both low-dose and high-dose XS markedly reduced late lumen loss resulting from vascular remodeling, as compared with the control group (P < 0.05 and P < 0.01 respectively). CONCLUSION: Vascular remodeling played an important role in late lumen stenosis after balloon injury of coronary artery. XS could significantly inhibit pathological vascular remodeling after balloon injury, thus reduce late lumen loss and prevent restenosis.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Coronary Restenosis/pathology , Coronary Vessels/pathology , Drugs, Chinese Herbal/pharmacology , Animals , Capsules , Coronary Restenosis/prevention & control , Female , Male , Phytotherapy , Swine
SELECTION OF CITATIONS
SEARCH DETAIL