Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Hum Hypertens ; 30(6): 392-6, 2016 06.
Article in English | MEDLINE | ID: mdl-26355832

ABSTRACT

Little is known about the potential progression of hypertensive patients towards isolated systolic hypertension (ISH) and about the phenotypes associated with the development of this condition. Aim of this study was to detect predictors of evolution towards ISH in patients with initial systolic-diastolic hypertension. We selected 7801 hypertensive patients free of prevalent cardiovascular (CV) diseases or severe chronic kidney disease and with at least 6-month follow-up from the Campania Salute Network. During 55±44 months of follow-up, incidence of ISH was 21%. Patients with ISH at the follow-up were significantly older (P<0.0001), had longer duration of hypertension, higher prevalence of diabetes and were more likely to be women (all P<0.0001). They exhibited higher baseline left ventricular mass index (LVMi), arterial stiffness (pulse pressure/stroke index), relative wall thickness (RWT) and carotid intima-media thickness (IMT; all P<0.001). Independent predictors of incident ISH were older age (odds ratio (OR)=1.14/5 years), female gender (OR=1.30), higher baseline systolic blood pressure (OR=1.03/5 mm Hg), lower diastolic blood pressure (OR=0.89/5 mm Hg), longer duration of hypertension (OR=1.08/5 months), higher LVMi (OR=1.02/5 g m(-2.7)), arterial stiffness (OR=2.01), RWT (OR=1.02), IMT (OR=1.19 mm(-1); all P<0.0001), independently of antihypertensive treatment, obesity, diabetes and fasting glucose (P>0.05). Our findings suggest that ISH is a sign of aggravation of the atherosclerotic disease already evident by the target organ damage. Great efforts should be paid to prevent this evolution and prompt aggressive therapy for arterial hypertension should be issued before the onset of target organ damage, to reduce global CV risk.


Subject(s)
Arterial Pressure , Hypertension/physiopathology , Adult , Aged , Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Carotid Artery Diseases/epidemiology , Chi-Square Distribution , Diastole , Disease Progression , Female , Humans , Hypertension/diagnosis , Hypertension/drug therapy , Hypertension/epidemiology , Hypertrophy, Left Ventricular/epidemiology , Italy/epidemiology , Logistic Models , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Phenotype , Prevalence , Prognosis , Registries , Risk Factors , Systole , Tertiary Care Centers , Vascular Stiffness
2.
J Cardiovasc Surg (Torino) ; 55(3): 335-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24755701

ABSTRACT

The treatment of in-stent restenosis (ISR) in the femoro-popliteal artery (FPA) is one of the major challenges of endovascular therapy, occurring in up to 40% of femoro-popliteal lesions treated with bare-metal stents within 1 year of treatment. Drug-eluting technologies, involving local delivery of paclitaxel, are providing a new paradigm for the treatment of ISR. Preliminary experience shows promising results compared to other techniques such as cutting balloon angioplasty and debulking strategies. Based on available data, drug-eluting balloons (DEBs) seem sufficient as stand-alone treatment of FPA-ISR. However, larger evidence from randomized studies is warranted to identify the clinical and/or anatomical setting in which they could fail.


Subject(s)
Angioplasty, Balloon/instrumentation , Cardiovascular Agents/administration & dosage , Femoral Artery , Peripheral Arterial Disease/therapy , Popliteal Artery , Vascular Access Devices , Angioplasty, Balloon/adverse effects , Animals , Constriction, Pathologic , Drug-Eluting Stents , Femoral Artery/diagnostic imaging , Femoral Artery/physiopathology , Humans , Neointima , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/physiopathology , Plaque, Atherosclerotic , Popliteal Artery/diagnostic imaging , Popliteal Artery/physiopathology , Prosthesis Design , Radiography , Recurrence , Retreatment , Time Factors , Treatment Outcome , Vascular Patency
3.
J Cardiovasc Surg (Torino) ; 54(1): 41-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23296414

ABSTRACT

Carotid artery stenting (CAS) is considered to be an acceptable alternative to carotid endarterectomy (CEA), particularly in patients at increased risk for CEA. The safety of CAS has improved significantly over the past decade. Adjunctive use of embolic protection devices (EPDs), is thought to be partially responsible for the improved outcomes. Among these, proximal EPDs are based on the surgical principle of stopping or reversing flow by clamping the carotid arteries, much in the same way carotid endarterectomy accomplishes neuroprotection. This is achieved through external and common carotid clamping. A recent meta-analysis demonstrates that the use of proximal EPDs for neuroprotection in patients undergoing CAS is associated with a very low incidence of any strokes and composite MACCE at 30 days. This study also demonstrates that the excellent outcomes achieved using proximal EPDs are independent of patient gender, symptomatic status, and other baseline clinical characteristics including the presence of a contralateral carotid occlusion.


Subject(s)
Carotid Arteries/surgery , Carotid Stenosis/surgery , Embolic Protection Devices , Stroke/prevention & control , Humans , Prosthesis Design , Stents
4.
Int J Cardiol ; 127(1): 98-102, 2008 Jun 23.
Article in English | MEDLINE | ID: mdl-18280596

ABSTRACT

BACKGROUND: Pulsed-wave ultraviolet excimer laser light at 308 nm can vaporise thrombus, suppress platelet aggregation, and, unlike other thrombectomy devices, ablates the underlying plaque. AIM: To evaluate both safety and efficacy of laser ablation in patients presenting with Acute Myocardial Infarction (AMI) complicated by persistent thrombotic occlusion. METHODS: From May 2003 to October 2006, we enrolled 66 AMI patients (age 59+/-11 years; 57 men) presenting complete thrombotic occlusion of the infarct related vessel. All patients were treated with laser. Primary acute angiographic end-points was corrected TIMI frame count. Secondary echocardiographic end-point was left ventricular remodeling defined as an increase in end-diastolic volume >/=20% 6 months after infarction. Tertiary clinical endpoint was event-free survival at 6 months follow-up. RESULTS: There were no intra-procedural death or coronary perforation. One primary angiographic failure was observed during lasing. Major dissection occurred in 1 (1.5%) and distal embolization in 4 patients (6%). Corrected TIMI frame count was 100 at baseline, 29+/-0.6 after lasing and 22+/-3 after stenting. At 6-months follow-up, left ventricular remodeling occurred in 8% patients. Event-free survival was 95% at 6-months follow-up. CONCLUSION: Laser angioplasty is feasible, safe and effective for the challenging treatment of patients with AMI and thrombus-laden lesions. The acute effects on coronary epicardial and myocardial reperfusion are excellent.


Subject(s)
Angioplasty, Balloon, Laser-Assisted , Coronary Thrombosis/surgery , Myocardial Infarction/surgery , Coronary Angiography , Coronary Thrombosis/complications , Coronary Thrombosis/diagnostic imaging , Echocardiography , Feasibility Studies , Female , Humans , Male , Middle Aged , Myocardial Infarction/complications , Myocardial Infarction/diagnostic imaging , Statistics, Nonparametric , Survival Analysis , Treatment Outcome
5.
Heart ; 94(2): 217-21, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17639100

ABSTRACT

OBJECTIVE: To verify whether optical coherence tomography (OCT) can accurately monitor the occurrence of arterial healing after stenting. SETTING: Delayed stent endothelialisation may predispose to stent thrombosis. OCT is a high-resolution intravascular imaging technique that accurately identifies stent struts and arterial tissues. DESIGN AND INTERVENTIONS: Eight New Zealand white rabbits underwent the implantation of single bare metal stents (diameter 2-2.5 mm, length 8-13 mm) in the right common carotid artery through the external carotid artery. After a median of 11 days (range 2-28), the stented arteries were visualised by OCT, with images acquired at a pull-back speed of 0.5 mm/sec. The rabbits were then euthanised, vessels were formalin-fixed and finally processed for histopathology. RESULTS: We analysed 32 cross-sections from eight stented carotid arteries, for a total of 384 stent struts. OCT detected all of the stent struts in 30 of 32 cross-sections (93.7%), and correctly identified the presence/absence of tissue for every strut. Histological and OCT measurements of mean neointima thickness (0.135 (SD 0.079) mm and 0.145 (SD 0.085) mm, respectively, p = NS) were similar and closely related (r = 0.85, p<0.001). Neointima area progressively increased with longer time intervals from stent deployment to sacrifice; histological and OCT measurements were similar for each time interval. The intra-observer and interobserver reproducibility of OCT neointima measurements were excellent (R2 = 0.90 and 0.88, respectively). CONCLUSIONS: OCT is a promising means for monitoring stent strut coverage and vessel wall healing in vivo, the relevance of which will become even more significant with the increasing use of drug-eluting stents.


Subject(s)
Carotid Artery Diseases/diagnostic imaging , Carotid Artery, Common/diagnostic imaging , Stents , Tomography, Optical Coherence , Animals , Carotid Artery Diseases/pathology , Carotid Artery Diseases/surgery , Carotid Artery, Common/pathology , Rabbits , Radiography , Tunica Intima/pathology , Wound Healing
6.
Circulation ; 109(12): 1543-9, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15023891

ABSTRACT

BACKGROUND: Bone marrow cell therapy is reported to contribute to collateral formation through cell incorporation into new or remodeling vessels. However, the possible role of a paracrine contribution to this effect is less well characterized. METHODS AND RESULTS: Murine marrow-derived stromal cells (MSCs) were purified by magnetic bead separation of cultured bone marrow. The release of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), placental growth factor (PlGF), and monocyte chemoattractant protein-1 (MCP-1) was demonstrated by analysis of MSC conditioned media (MSC-CM). MSC-CM enhanced proliferation of endothelial cells and smooth muscle cells in a dose-dependent manner; anti-VEGF and anti-FGF antibodies only partly attenuated these effects. Balb/C mice (n=10) underwent distal femoral artery ligation, followed by adductor muscle injection of 1x10(6) MSCs 24 hours later. Compared with controls injected with media (n=10) or mature endothelial cells (n=8), distal limb perfusion improved, and mid-thigh conductance vessels increased in number and total cross-sectional area. MSC injection improved limb function and appearance, reduced the incidence of auto-amputation, and attenuated muscle atrophy and fibrosis. After injection, labeled MSCs were seen dispersed between muscle fibers but were not seen incorporated into mature collaterals. Injection of MSCs increased adductor muscle levels of bFGF and VEGF protein compared with controls. Finally, colocalization of VEGF and transplanted MSCs within adductor tissue was demonstrated. CONCLUSIONS: MSCs secrete a wide array of arteriogenic cytokines. MSCs can contribute to collateral remodeling through paracrine mechanisms.


Subject(s)
Collateral Circulation , Growth Substances/metabolism , Hindlimb/blood supply , Ischemia/therapy , Mesenchymal Stem Cell Transplantation , Paracrine Communication , Stromal Cells/transplantation , Animals , Cells, Cultured/metabolism , Cells, Cultured/transplantation , Chemokine CCL2/metabolism , Culture Media, Conditioned/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/metabolism , Fibrosis , Hypoxia-Inducible Factor 1, alpha Subunit , Immunomagnetic Separation , Injections, Intramuscular , Ischemia/physiopathology , Mice , Mice, Inbred BALB C , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Placenta Growth Factor , Pregnancy Proteins/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Circ Res ; 94(5): 678-85, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14739163

ABSTRACT

We recently demonstrated that marrow stromal cells (MSCs) augment collateral remodeling through release of several cytokines such as VEGF and bFGF rather than via cell incorporation into new or remodeling vessels. The present study was designed to characterize the full spectrum of cytokine genes expressed by MSCs and to further examine the role of paracrine mechanisms that underpin their therapeutic potential. Normal human MSCs were cultured under normoxic or hypoxic conditions for 72 hours. The gene expression profile of the cells was determined using Affymetrix GeneChips representing 12 000 genes. A wide array of arteriogenic cytokine genes were expressed at baseline, and several were induced >1.5-fold by hypoxic stress. The gene array data were confirmed using ELISA assays and immunoblotting of the MSC conditioned media (MSC(CM)). MSC(CM) promoted in vitro proliferation and migration of endothelial cells in a dose-dependent manner; anti-VEGF and anti-FGF antibodies only partially attenuated these effects. Similarly, MSC(CM) promoted smooth muscle cell proliferation and migration in a dose-dependent manner. Using a murine hindlimb ischemia model, murine MSC(CM) enhanced collateral flow recovery and remodeling, improved limb function, reduced the incidence of autoamputation, and attenuated muscle atrophy compared with control media. These data indicate that paracrine signaling is an important mediator of bone marrow cell therapy in tissue ischemia, and that cell incorporation into vessels is not a prerequisite for their effects.


Subject(s)
Blood Vessels/growth & development , Bone Marrow Cells/physiology , Collateral Circulation/physiology , Cytokines/physiology , Gene Expression Profiling , Paracrine Communication , Adult , Animals , Blood Vessels/cytology , Cell Division/drug effects , Cell Hypoxia , Cell Movement/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Collateral Circulation/drug effects , Culture Media, Conditioned/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Cytokines/therapeutic use , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Female , Gene Expression Regulation , Humans , Ischemia/drug therapy , Ischemia/physiopathology , Mice , Mice, Inbred BALB C , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscular Atrophy/etiology , Muscular Atrophy/prevention & control , Myocytes, Smooth Muscle/drug effects , Stromal Cells/cytology , Stromal Cells/metabolism
8.
Minerva Cardioangiol ; 50(6): 637-42, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12473983

ABSTRACT

Troponin measurement has now become an integral part of the assessment of patients with acute coronary syndromes (ACS). Obtaining troponin levels have been used effectively as a diagnostic tool with superior sensitivity and specificity compared to creatine kinase MB fraction in identifying high-risk ACS patients. The adverse prognosis of these high-risk patients can be modified by more aggressive treatment strategy including antiplatelet and antithrombotic therapy accompanied, if feasible, by early percutaneous intervention. The current review summarizes available data on troponin measurement as a clinical tool for tailoring therapeutic strategy in non-ST elevation ACS patients. In addition, the data on the potential predictive value of postintervention troponin levels is discussed.


Subject(s)
Coronary Disease/blood , Troponin/blood , Acute Disease , Cardiology , Coronary Angiography , Coronary Disease/diagnostic imaging , Coronary Disease/therapy , Humans
9.
Catheter Cardiovasc Interv ; 54(4): 533-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11747197

ABSTRACT

Early studies have indicated no correlation between the amount of mechanical injury and the level of myocardial gene expression following direct plasmid vector injection. Recently, however, evidence suggests that combined laser myocardial injury and plasmid-based gene delivery exert synergistic effects on gene expression and activity. The purpose of the study was to determine whether laser-induced myocardial injury followed by transendocardial gene transfer increases gene expression compared to gene transfer alone. We assessed the ability of a plasmid vector to express its transgene after injection into porcine ischemic myocardium with and without preceding laser myocardial injury. Thirteen animals had transendocardial injections of the luciferase reporter gene in a plamid vector using a catheter-based injection system. Injections (0.5 mg per animal, 50 microg per injection site) were divided into 10 sites in the ischemic territory. Eight animals underwent transendocardial laser injury of the ischemic region (2 Joule per pulse x 10 sites) prior to gene delivery. In five animals, gene injection sites were dispersed between laser channels, and in three animals laser and gene delivery were applied in close proximity (< 5 mm) or at the same location. Luciferase activity was measured at 3 and 7 days. Luciferase expression in ischemic zones was markedly elevated at day 3 and 7, and similar whether animals were pretreated using laser injury followed by gene transfer compared to gene transfer alone. Neither same-spot injection nor dispersed gene delivery were associated with augmented gene expression compared to gene transfer alone. Using the above-described catheter-based approach to combine localized laser injury and injection of naked DNA into ischemic myocardium, laser injury did not augment gene expression above levels present with gene transfer alone.


Subject(s)
Genetic Therapy/methods , Heart Injuries/etiology , Laser Therapy , Myocardial Ischemia/genetics , Myocardial Ischemia/surgery , Myocardium/metabolism , Plasmids/genetics , Plasmids/therapeutic use , Animals , Electrophysiologic Techniques, Cardiac , Gene Expression/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Heart Ventricles/injuries , Luciferases/biosynthesis , Luciferases/genetics , Models, Cardiovascular , Swine , Treatment Outcome
10.
Arterioscler Thromb Vasc Biol ; 21(10): 1604-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11597933

ABSTRACT

The clinical outcome of vascular stenting is limited by in-stent stenosis. Increased nitric oxide (NO)/cGMP signaling by L-arginine (L-Arg) supplementation, the substrate for NO synthase (NOS), or NOS gene transfer may reduce in-stent neointima formation. After stenting, vascular cell proliferation in rat carotid arteries, as measured by 5'-bromodeoxyuridine (5'-BrdU) incorporation, indicated 15+/-8%, 28+/-5%, and 33+/-7% 5'-BrdU-positive vascular cells at 4, 7, and 14 days, respectively. Reporter beta-galactosidase gene transfer efficacy was evidenced by 30% beta-galactosidase-expressing medial smooth muscle cells at 14 days. The intima-to-media ratio (I/M) progressively increased to 2.32+/-0.24 at 14 days. To target in-stent neointima formation, animals were infected with adenoviral vectors (4x10(10) plaque-forming units per mL) expressing NOS2 (AdNOS2) or no transgene (AdRR5), or they received daily doses of L-Arg (500 mg. kg(-1). (d-1) IP). The neointima at 14 days was smaller in L-Arg-treated than in untreated rats (I/M 1.25+/-0.35 vs 2.32+/-0.24, P<0.05, n=7 each) or in AdRR5- and AdNOS2-infected rats (I/M 2.57+/-0.43, n=7 and 1.82+/-0.75, n=8, respectively; P<0.05 for both). The effect of L-Arg was abolished by simultaneous administration of N(G)-nitro L-arginine methyl ester, an NOS inhibitor (2.03+/-0.39, P<0.05, vs L-Arg). Inflammation was markedly less in L-Arg- and AdNOS2-treated than in AdRR5-infected rats. Supplemental L-Arg reduces neointima formation after stenting by way of an NOS-dependent mechanism and may be a valuable strategy to target in-stent stenosis.


Subject(s)
Arginine/pharmacology , Carotid Stenosis/therapy , Nitric Oxide/physiology , Stents/adverse effects , Adenoviridae/genetics , Angioplasty, Balloon/adverse effects , Animals , Arginine/administration & dosage , Carotid Stenosis/etiology , Carotid Stenosis/pathology , Genetic Therapy , Genetic Vectors , Male , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Transduction, Genetic
11.
Am J Cardiol ; 88(2): 129-33, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11448408

ABSTRACT

Troponin I is a predictive marker of short- and intermediate-term adverse cardiac events in patients with acute coronary syndromes (ACS). These high-risk patients may benefit from early percutaneous coronary intervention. However, whether additional myocardial injury, defined as postprocedural troponin I elevation, may be associated with adverse short- and intermediate-term outcomes has not been fully explored. Accordingly, we studied 132 consecutive patients with non-ST-elevation ACS (62% with non-Q-wave myocardial infarction) and elevated troponin I levels at admission (>0.15 ng/ml) who underwent percutaneous coronary intervention > or =48 hours after admission. Troponin I levels were routinely measured at 6 and 18 to 24 hours after intervention and patients were stratified according to the presence or absence of troponin I re-elevation, defined as postprocedural troponin I levels >1 times the admission levels. In-hospital and cumulative 6-month clinical outcomes were compared between groups. Patients with troponin I re-elevation (n = 51) were older (68 +/- 13 vs 64 +/- 12 years, p = 0.05) and had experienced prior myocardial infarction more frequently (92.5 vs 82.1, p = 0.09), but otherwise had similar baseline clinical characteristics. Patients with troponin I re-elevation had significantly higher in-hospital mortality (9.8% vs 0%, p = 0.016) and a higher 6-month cumulative death rate (24% vs 3.7%, p = 0.001). There was a trend for an increased 6-month myocardial infarction rate in patients with troponin I re-elevation (13.7% vs 3.7%, p = 0.11) and target vessel revascularization was similar between groups (16.7% vs 17.4%, p = 0.92). By multivariate analysis, troponin I re-elevation (odds ratio [OR] 6.2, p = 0.011) and diabetes mellitus (OR 5.7, p = 0.014) were the strongest independent predictors for increased 6-month cumulative mortality, whereas creatine kinase MB-fraction re-elevation had no prognostic value. We conclude that troponin I re-elevation after percutaneous coronary intervention in high-risk patients with ACS is associated with a substantial increase in mortality and reduced event-free survival at 6-month follow-up.


Subject(s)
Myocardial Infarction/blood , Myocardial Infarction/therapy , Troponin I/blood , Aged , Angioplasty, Balloon, Coronary , Cohort Studies , Creatine Kinase/blood , Creatine Kinase, MB Form , Disease-Free Survival , Female , Follow-Up Studies , Hospital Mortality , Humans , Isoenzymes/blood , Male , Middle Aged , Myocardial Infarction/mortality , Predictive Value of Tests , Prognosis , Risk Assessment , Stents , Time Factors
12.
Circulation ; 103(24): 2980-6, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11413090

ABSTRACT

BACKGROUND: The mechanisms of increased neointimal hyperplasia after coronary interventions in diabetic patients are still unknown. METHODS AND RESULTS: Glucose and insulin effects on in vitro vascular smooth muscle cell (VSMC) proliferation and migration were assessed. The effect of balloon injury on neointimal hyperplasia was studied in streptozotocin-induced diabetic rats with or without adjunct insulin therapy. To study the effect of balloon injury in nondiabetic rats with hyperinsulinemia, pancreatic islets were transplanted under the kidney capsule in normal rats. Glucose did not increase VSMC proliferation and migration in vitro. In contrast, insulin induced a significant increase in VSMC proliferation and migration in cell cultures. Furthermore, in VSMC culture, insulin increased MAPK activation. A reduction in neointimal hyperplasia was consistently documented after vascular injury in hyperglycemic streptozotocin-induced diabetic rats. Insulin therapy significantly increased neointimal hyperplasia in these rats. This effect of hyperinsulinemia was totally abolished by transfection on the arterial wall of the N17H-ras-negative mutant gene. Finally, after experimental balloon angioplasty in hyperinsulinemic nondiabetic islet-transplanted rats, a significant increase in neointimal hyperplasia was observed. CONCLUSIONS: In rats with streptozotocin-induced diabetes, balloon injury was not associated with an increase in neointimal formation. Exogenous insulin administration in diabetic rats and islet transplantation in nondiabetic rats increased both blood insulin levels and neointimal hyperplasia after balloon injury. Hyperinsulinemia through activation of the ras/MAPK pathway, rather than hyperglycemia per se, seems to be of crucial importance in determining the exaggerated neointimal hyperplasia after balloon angioplasty in diabetic animals.


Subject(s)
Angioplasty, Balloon , Carotid Artery Diseases/pathology , Diabetes Mellitus, Experimental/pathology , Hyperinsulinism/pathology , Hyperplasia/pathology , Islets of Langerhans Transplantation , Tunica Intima/pathology , Angioplasty, Balloon/adverse effects , Animals , Blood Glucose , Carotid Artery Diseases/etiology , Carotid Artery Diseases/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Movement/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Glucose/pharmacology , Hyperinsulinism/chemically induced , Hyperinsulinism/metabolism , Hyperplasia/etiology , Hyperplasia/genetics , Insulin/blood , Insulin/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Mutagenesis, Site-Directed , Rats , Rats, Inbred F344 , Rats, Wistar , Signal Transduction/drug effects , Streptozocin , Transfection , Tunica Intima/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/genetics
13.
J Am Coll Cardiol ; 37(6): 1639-44, 2001 May.
Article in English | MEDLINE | ID: mdl-11345378

ABSTRACT

OBJECTIVES: We tested the hypothesis that the response to flecainide infusion can identify patients with atrial fibrillation (AF) in whom the hybrid pharmacologic and ablation therapy reduces the recurrences of AF. BACKGROUND: Infusion of class IC anti-arrhythmic drugs may promote transformation of AF into atrial flutter. Catheter ablation of atrial flutter has been demonstrated to be highly effective in preventing recurrences of atrial flutter. METHODS: Seventy-one consecutive patients with paroxysmal or chronic AF, in whom flecainide infusion (2 mg/kg body weight, intravenously) determined the transformation of AF into common atrial flutter (positive response), were randomized to receive one of the following treatments: oral pharmacologic treatment with flecainide (group A, n = 23); the hybrid treatment (catheter ablation of the inferior vena cava-tricuspid annulus isthmus, plus oral flecainide) (group B, n = 24); or catheter ablation of the isthmus only (group C, n = 24). Thirty-seven patients with a negative response to flecainide, who chose to be submitted to the hybrid treatment, were selected as the control group (group D). RESULTS: During a mean follow-up period of 24 +/- 7.2 months, the recurrences of AF and atrial flutter in group B (42%) were significantly lower than those in group A (78%, p < 0.001), group C (92%, p < 0.001) and group D (92%, p < 0.001). CONCLUSIONS: The creation of a complete bi-directional conduction block at the inferior vena cava-tricuspid annulus isthmus, plus flecainide administration, reduces the recurrences of both AF and atrial flutter in patients with class IC atrial flutter. Moreover, the early response to flecainide is safe and reliable in identifying patients who may benefit from this therapy.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Catheter Ablation , Flecainide/therapeutic use , Administration, Oral , Aged , Analysis of Variance , Atrial Fibrillation/complications , Atrial Fibrillation/diagnosis , Atrial Flutter/etiology , Catheter Ablation/methods , Chronic Disease , Combined Modality Therapy , Disease-Free Survival , Electrocardiography , Female , Follow-Up Studies , Humans , Infusions, Intravenous , Male , Middle Aged , Patient Selection , Predictive Value of Tests , Proportional Hazards Models , Recurrence , Treatment Outcome
14.
Circ Res ; 88(3): 319-24, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11179200

ABSTRACT

cAMP-dependent protein kinase is anchored to discrete cellular compartments by a family of proteins, the A-kinase anchor proteins (AKAPs). We have investigated in vivo and in vitro the biological effects of the expression of a prototypic member of the family, AKAP75, on smooth muscle cells. In vitro expression of AKAP75 in smooth muscle cells stimulated cAMP-induced transcription, increased the levels of the cyclin-dependent kinase-2 inhibitor p27(kip1), and reduced cell proliferation. In vivo expression of exogenous AKAP75 in common carotid arteries, subjected to balloon injury, significantly increased the levels of p27(kip1) and inhibited neointimal hyperplasia. Both the effects in smooth muscle cells in vitro and in carotid arteries in vivo were specifically dependent on the amplification of cAMP-dependent protein kinase (PKA) signals by membrane-bound PKA, as indicated by selective loss of the AKAP75 biological effects in mutants defective in the PKA anchor domain or by suppression of AKAP effects by the PKA-specific protein kinase inhibitor. These data indicate that AKAP proteins selectively amplify cAMP-PKA signaling in vitro and in vivo and suggest a possible target for the inhibition of the neointimal hyperplasia after vascular injury.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Division/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Tumor Suppressor Proteins , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , A Kinase Anchor Proteins , Animals , Carotid Arteries/chemistry , Carotid Arteries/pathology , Carrier Proteins/genetics , Cell Division/drug effects , Cells, Cultured , Chloramphenicol O-Acetyltransferase/drug effects , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclin-Dependent Kinase Inhibitor p27 , DNA/biosynthesis , DNA/drug effects , DNA, Recombinant , Gene Transfer Techniques , Immunohistochemistry , Microtubule-Associated Proteins/analysis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Plasmids/genetics , Rats , Rats, Wistar , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Time Factors , Tunica Intima/chemistry , Tunica Intima/pathology , Tunica Media/chemistry , Tunica Media/pathology
15.
J Am Coll Cardiol ; 36(1): 288-93, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898448

ABSTRACT

OBJECTIVES: The aims of the present study were to assess 1) the effect of 8-C1-cAMP (cyclic-3'-5'-adenosine monophosphate) on vascular smooth muscle cell (VSMC) proliferation in vitro and 2) the efficacy of systemic administration of 8-C1-cAMP on neointimal formation after balloon injury in vivo. BACKGROUND: Neointimal formation after vascular injury is responsible for restenosis after arterial stenting. Recently, 8-C1-cAMP, a cAMP analogue that induces growth arrest, has been safely administered in phase I studies in humans. METHODS: The effect of 8-C1-cAMP on cell proliferation was first assessed on SMCs in vitro. To study the effects of cAMP in vivo, balloon injury was performed in 67 rats using a 2F Fogarty balloon catheter. RESULTS: The 8-C1-cAMP markedly inhibited VSMC proliferation in vitro, reduced protein kinase A (PKA) RIalpha subunit expression, and induced PKA RIIbeta subunit expression. In addition, 8-C1-cAMP reduced, in a dose-dependent manner, neointimal area and neointima/media ratio after balloon injury. The proliferative activity, assessed by proliferating nuclear cell antigen immunostaining, revealed a reduction of proliferative activity of VSMCs in vivo in the 8-C1-cAMP group. Moreover, the systemic administration of 8-C1-cAMP did not affect renal function, blood pressure and heart rate. CONCLUSIONS: We conclude that 8-C1-cAMP potently inhibits VSMC proliferation in vitro and reduces neointima formation by balloon injury in vivo after systemic administration. These data may have a clinical relevance in designing future strategies to prevent restenosis after arterial stenting and perhaps after percutaneous transluminal coronary angioplasty.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , Antineoplastic Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Tunica Intima/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/administration & dosage , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Aorta, Thoracic/injuries , Aorta, Thoracic/pathology , Arterial Occlusive Diseases/enzymology , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/prevention & control , Blood Pressure/drug effects , Catheterization/adverse effects , Cell Division/drug effects , Cells, Cultured , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation/drug effects , Heart Rate/drug effects , Injections, Intraperitoneal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Rats , Rats, Wistar , Tunica Intima/cytology , Tunica Intima/enzymology , Up-Regulation/drug effects
16.
Basic Res Cardiol ; 95(3): 179-85, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10879619

ABSTRACT

OBJECTIVES: Restenosis is the major complication of coronary angioplasty and stenting. In addition, the small vessel diameter represents a major limitation to the wide use of the technology. The aim of this study was to assess the feasibility and the vascular response of stent deployment in rat small vessels. METHODS: In 40 Wistar rats (500-550 g) a Nir stent crimped on a 1.5 mm Comet angioplasty balloon catheter was deployed at high pressure in the common carotid artery. Neointimal area, neointima/media ratio and the arterial dimension were assessed immediately and at 7, 14, 21, and 28 days after stenting. RESULTS: After stent deployment, the neointimal area and the neointima/media ratio increased progressively and peaked at 14 days (p < 0.05 vs 0 and 7 days). Alpha-actin-positive cells were found circumferentially organized on the lumen surface. At 21 and 28 days after stenting, the neointima and the neointima/media ratio were not statistically different compared with the results obtained fourteen days after stent deployment. No significant differences in the area of external elastic lamina were observed during the study period. In contrast, the internal lumen area was reduced significantly at 14, 21, and 28 days after the stent deployment. Subacute thrombosis rate after stent implantation was 26.5 %. CONCLUSIONS: The results of this study demonstrated that the balloon expandable stents can be safely placed into rat arteries and the reduction of the internal arterial lumen observed after stent deployment was only due to the neointima formation whereas remodeling did not occur.


Subject(s)
Carotid Artery, Common , Stents , Animals , Carotid Artery Diseases/etiology , Carotid Artery, Common/pathology , Catheterization , Feasibility Studies , Rats , Rats, Wistar , Stents/adverse effects , Thrombosis/etiology , Tunica Intima/pathology , Tunica Media/pathology
17.
J Am Coll Cardiol ; 35(1): 214-21, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10636283

ABSTRACT

OBJECTIVES: We sought to evaluate the effects of hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase inhibitors on vascular smooth muscle cell (VSMC) proliferation in vitro and neointimal formation in vivo after vascular injury. BACKGROUND: Neointimal hyperplasia after vascular injury is responsible for restenosis after arterial stenting, whereas arterial remodeling and neointimal formation are the causes of restenosis after percutaneous transluminal coronary angioplasty. METHODS: We assessed the effect of simvastatin on in vitro VSMC proliferation. To study the effects of simvastatin in vivo, balloon injury and stent deployment were performed in the common carotid artery of rats. Neointimal area was measured two weeks later in the balloon injury model and three weeks after stent deployment. RESULTS: Simvastatin markedly inhibits VSMC proliferation in vitro. In vivo, simvastatin reduced, in a dose-dependent manner, the neointimal area and the neointima-media ratio after balloon injury from 0.266 +/- 0.015 mm2 to 0.080 +/- 0.026 mm2 and from 1.271 +/- 0.074 to 0.436 +/- 0.158 (p < 0.001 vs. control rats) at the highest dose. Simvastatin also significantly reduced the neointimal formation and the neointima-media ratio after stenting from 0.508 +/- 0.035 mm2 to 0.362 +/- 0.047 mm2 (p < 0.05 vs. control rats) and from 2.000 +/- 0.136 to 1.374 +/- 0.180 (p < 0.05 vs. control rats). The vessel thrombosis rate after stent deployment was 30% in the control group and 11.1% in the treated group (p = NS). Moreover, the systemic administration of simvastatin did not affect hepatic and renal functions, blood pressure or heart rate. CONCLUSIONS: Simvastatin potently inhibits VSMC proliferation in vitro and reduces neointimal formation in a rat model of vascular injury.


Subject(s)
Cell Division/drug effects , Graft Occlusion, Vascular/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Muscle, Smooth, Vascular/drug effects , Simvastatin/pharmacology , Stents , Tunica Intima/drug effects , Animals , Cell Division/physiology , Cells, Cultured , In Vitro Techniques , Male , Muscle, Smooth, Vascular/pathology , Rats , Rats, Wistar , Recurrence , Tunica Intima/pathology , Tunica Media/drug effects , Tunica Media/pathology , Wound Healing/drug effects , Wound Healing/physiology
18.
J Biol Chem ; 274(10): 6546-52, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10037748

ABSTRACT

cAMP signals are received and transmitted by multiple isoforms of cAMP-dependent protein kinases, typically determined by their specific regulatory subunits. In the brain the major regulatory isoform RIIbeta and the RII-anchor protein, AKAP150 (rat) or 75 (bovine), are differentially expressed. Cortical neurons express RIIbeta and AKAP75; conversely, granule cerebellar cells express predominantly RIalpha and RIIalpha. Cortical neurons accumulate PKA catalytic subunit and phosphorylated cAMP responsive element binding protein very efficiently into nuclei upon cAMP induction, whereas granule cerebellar cells fail to do so. Down-regulation of RIIbeta synthesis by antisense oligonucleotides inhibited cAMP-induced nuclear signaling in cortical neurons. Expression in cerebellar granule cells of RIIbeta and AKAP75 genes by microinjection of specific expression vectors, markedly stimulated cAMP-induced transcription of the lacZ gene driven by a cAMP-responsive element promoter. These data indicate that the composition of PKA in cortical and granule cells underlies the differential ability of these cells to transmit cAMP signals to the nucleus.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins , Cell Nucleus/metabolism , Cerebellum/metabolism , Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Proteins/metabolism , Signal Transduction , A Kinase Anchor Proteins , Animals , Cerebellum/cytology , Cerebral Cortex/cytology , Neurons/metabolism , Neurons/ultrastructure , Protein Isoforms/metabolism , Rats
20.
Int J Mol Med ; 2(2): 143-148, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9855680

ABSTRACT

Angioplasty is a principal treatment for occlusive vascular disease but 30-50% of patients show a restenosis of the vessel. There is no clinical effective therapy for this disease. It has been demonstrated, in animal models, that various drugs such as NO-donor, plasminogen inhibitor tranexamic acid and MMP (matrix metalloproteinases) reduce the rate of restenosis. Other therapeutic approaches are cytotoxic therapy, and strategies to inhibit cell cycle progression. Systemic administration of conventional pharmacologic agents inhibit cell cycle kinases and vascular lesion formation in animal models. As cell cycle progression is accompanied by fluctuations in the concentration of adenosine 3',5-monophospate (cAMP) and in the activity of the cAMP dependent protein kinase (PKA), local administration of cAMP and phospodiesterase-inhibitor drugs (aminophylline or amrinone) markedly inhibit neointima formation. The successful use of local radiation therapy to inhibit neointima formation after vascular injury may reflect a similar combination of cell-cycle arrest and vascular cell apoptosis. The most effective therapy for occlusive vascular disease will likely combine intravascular stenting with an antiproliferative therapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...