Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Circulation ; 103(20): 2514-20, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11369694

ABSTRACT

BACKGROUND: The contribution of nitric oxide synthase (NOS)-2 to myocardial inflammation and cardiomyocyte necrosis and apoptosis during allograft rejection was investigated through heterotopic cardiac transplantation in mice. METHODS AND RESULTS: In the first experiments, hearts from C3H donor mice were transplanted into NOS-2(-/-) and NOS-2(+/+) C57BL/6J.129J recipients. A second series of experiments included NOS-2(-/-) donor hearts transplanted into NOS-2(-/-) recipients and wild-type NOS-2(+/+) donor hearts transplanted into wild-type NOS-2(+/+) recipients. (All donors were C57BL/6J and recipients were C57BL/6J.129J.) In the first series of experiments, no significant differences were observed in allograft survival, rejection score, total number of apoptotic nuclei (TUNEL), total number of apoptotic cardiomyocytes, or graft NOS-2 mRNA and protein. Positive NOS-2 immunostaining occurred in endothelial cells and cardiomyocytes in the allografts; the inflammatory infiltrate was NOS-2 positive only when recipients were NOS-2(+/+). In the second series of experiments, cardiac allograft survival was significantly increased in the NOS-2(-/-) mice (26+/-13 versus 17+/-8 days, P<0.05), along with significant reductions in inflammatory infiltrate, rejection score, and total number of apoptotic nuclei (23.5+/-9.5 versus 56.4+/-15.3, P<0.01) and of apoptotic cardiomyocytes (2.9+/-1.6 versus 6.9+/-2.7, P<0.05). No NOS-2 or nitrotyrosine, a marker of peroxynitrite exposure, was detected in NOS-2(-/-) allografts transplanted into NOS-2(-/-) recipients. CONCLUSIONS: The data suggest that NO derived from NOS-2 contributes to the inflammatory response and to cardiomyocyte damage and apoptosis during acute cardiac allograft rejection.


Subject(s)
Graft Rejection/enzymology , Heart Transplantation , Nitric Oxide Synthase/genetics , Acute Disease , Animals , Apoptosis , Female , Genotype , Graft Rejection/pathology , In Situ Nick-End Labeling , Inflammation/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Time Factors , Transplantation, Homologous
2.
J Am Coll Cardiol ; 37(1): 109-16, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11153724

ABSTRACT

OBJECTIVE: The study was done to determine whether coronary steal (defined as an absolute decrease in perfusion from resting blood flow) is induced by intravenous (IV) dipyridamole in patients with severe coronary artery disease (CAD). BACKGROUND: Myocardial ischemia during coronary vasodilation is usually attributed to coronary steal. However, there is limited data on the absolute magnitude of coronary steal in humans. METHODS: Eighteen patients with multivessel CAD underwent dynamic positron emission tomography (PET) imaging with 13NH3 at rest and after infusion of IV dipyridamole. Eight myocardial sectors were analyzed per short axis slice and myocardial blood flow calculated with a two-compartment model in absolute terms. RESULTS: Coronary steal occurred in 8 of the 18 patients. In the 8 patients with coronary steal, myocardial blood flow decreased from 90 +/- 18 ml/100 g/min at rest to 68 +/- 27 ml/100 g/min following dipyridamole in the segments with steal, and increased from 87 +/- 19 to 138 +/- 16 ml/100 g/min following dipyridamole in the segments without steal. Significant clinical correlates of coronary steal were either ST elevation or the combination of ST depression and angina. CONCLUSIONS: Coronary vasodilation with IV dipyridamole is associated with significant reductions in blood flow to collateral-dependent myocardium consistent with coronary steal in about 45% of patients with severe CAD.


Subject(s)
Coronary Circulation/drug effects , Coronary Disease/diagnosis , Dipyridamole , Myocardial Ischemia/diagnosis , Aged , Collateral Circulation/drug effects , Collateral Circulation/physiology , Coronary Circulation/physiology , Coronary Disease/physiopathology , Female , Humans , Image Processing, Computer-Assisted , Infusions, Intravenous , Male , Middle Aged , Myocardial Ischemia/physiopathology , Tomography, Emission-Computed , Vasodilation/drug effects , Vasodilation/physiology
3.
Virchows Arch ; 437(2): 149-59, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10993275

ABSTRACT

T cells have roles in the pathogenesis of native coronary atherosclerosis (CA) and transplant-associated coronary artery disease (TCAD). The mechanisms by which T cells interact with other cells in these lesions are not fully known. CD154 is an activation-induced CD4+ T cell surface molecule that interacts with CD40+ target cells, including macrophages and endothelial cells, and induces the production of pro-inflammatory molecules, including CD54 (ICAM-1) and CD106 (VCAM-1). To investigate whether CD154-CD40 interactions might be involved in the pathogenesis of CA or TCAD we performed immunohistochemical studies of CD154 and CD40 expression on frozen sections of coronary arteries obtained from cardiac allograft recipients with CA (n=10) or TCAD (n=9). Utilizing four different anti-CD154 mAb we found that CD154 expression was restricted to infiltrating lymphocytes in CA and TCAD. CD40 expression was markedly up-regulated on intimal endothelial cells, foam cells, macrophages and smooth muscle cells in both diseases. Dual immunolabeling demonstrated many CD40+ cells co-expressed CD54 and CD106. The extent of CD40, CD54 and CD106 expression showed statistical significant correlation with the severity of disease and the amount of intimal lymphocytes. Together these studies demonstrate the presence of activated CD154+ and CD40+ cells in both CA and TCAD lesions and suggest that CD154-mediated interactions with CD40+ macrophages, foam cells, smooth muscle cells and/or endothelial cells may contribute to the pathogenesis of these diseases.


Subject(s)
CD40 Antigens/metabolism , Coronary Artery Disease/metabolism , Coronary Disease/metabolism , Coronary Vessels/metabolism , Heart Transplantation , Membrane Glycoproteins/metabolism , Postoperative Complications/metabolism , CD40 Ligand , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Immunohistochemistry , Reference Values
4.
Arterioscler Thromb Vasc Biol ; 20(9): 2039-44, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10978246

ABSTRACT

Asymmetric dimethylarginine (ADMA), a compound detectable in human plasma, is an endogenous inhibitor of NO synthase. Endothelial dysfunction is an early event in atherogenesis, and large-vessel atherosclerosis is a major cause of morbidity and mortality in patients with type 2 diabetes mellitus. Fifty patients with type 2 diabetes mellitus were studied at baseline and 5 hours after ingestion of a high-fat meal. Plasma ADMA measured by using high-performance liquid chromatography increased from 1.04+/-0.99 to 2.51+/-2.27 micromol/L (P:<0.0005). Brachial arterial vasodilation after reactive hyperemia, a NO-dependent function, measured by high-resolution ultrasound, decreased from 6.9+/-3.9% at baseline to 1.3+/-4.5% (P:<0.0001). These changes occurred in association with increased plasma levels of triglycerides and very low density lipoprotein triglycerides, with reduced low density lipoprotein cholesterol and high density lipoprotein cholesterol, and with no changes in total cholesterol. The increase in plasma ADMA in response to a high-fat meal was significantly and inversely related to the decrease in percent vasodilation. In 10 of the subjects studied with a similar protocol on another day, no significant changes in the brachial artery flow responses or in plasma ADMA were observed 5 hours after ingestion of a nonfat isocaloric meal. The data suggest that ADMA may contribute to abnormal blood flow responses and to atherogenesis in type 2 diabetics.


Subject(s)
Arginine/analogs & derivatives , Arginine/blood , Diabetes Mellitus, Type 2/blood , Dietary Fats/pharmacology , Endothelium, Vascular/drug effects , Adult , Aged , Endothelium, Vascular/physiology , Female , Humans , Lipids/blood , Male , Middle Aged , Triglycerides/blood , Vasodilation/drug effects
5.
Circulation ; 101(4): 430-8, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10653836

ABSTRACT

BACKGROUND: The hypothesis that cyclooxygenase-2 (COX-2) is involved in the myocardial inflammatory response during cardiac allograft rejection was investigated using a rat heterotopic abdominal cardiac transplantation model. METHODS AND RESULTS: COX-2 mRNA and protein in the myocardium of rejecting cardiac allografts were significantly elevated 3 to 5 days after transplantation compared with syngeneic controls (n=3, P<0.05). COX-2 upregulation paralleled in time and extent the upregulation of iNOS mRNA, protein, and enzyme activity in this model. COX-2 immunostaining was prominent in macrophages infiltrating the rejecting allografts and in damaged cardiac myocytes. Prostaglandin (PG) levels in rejecting allografts were also higher than in native hearts. Because NO has been reported to modulate PG synthesis by COX-2, additional transplants were performed using animals treated with a selective COX-2 inhibitor (SC-58125) and a selective inhibitor of the inducible nitric oxide synthase (iNOS) N-aminomethyl-L-lysine. At posttransplant day 5, inhibitor administration resulted in a significant reduction of COX-2 mRNA expression (3764+/-337 versus 5110+/-141 arbitrary units, n=3, P<0.05) and iNOS enzymatic activity (1.7+/-0.4 versus 22.8+/-14. 4 nmol/mg protein, n=3, P<0.01) compared with vehicle-treated allogeneic transplants. Allograft survival in treated animals was increased modestly from 5.4 to 6.4 days (P<0.05). However, apoptosis of cardiac myocytes (TUNNEL method) was only marginally reduced relative to vehicle controls in treated graft recipients. The intensity of allograft rejection was also similar in the treated and untreated allografts. CONCLUSIONS: The data indicates that COX-2 expression is enhanced in parallel with iNOS in the myocardium during cardiac allograft rejection.


Subject(s)
Gene Expression Regulation, Enzymologic , Graft Rejection/enzymology , Heart Transplantation/immunology , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Animals , Cyclooxygenase 2 , Graft Rejection/pathology , Heart Transplantation/pathology , Isoenzymes/metabolism , Male , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Rats, Inbred WF , Time Factors , Transcription, Genetic , Transplantation, Heterotopic , Transplantation, Homologous , Transplantation, Isogeneic
7.
Am J Physiol ; 277(3): H1189-99, 1999 09.
Article in English | MEDLINE | ID: mdl-10484441

ABSTRACT

Excessive nitric oxide (NO) production within the heart is implicated in the pathogenesis of myocyte death, but the mechanism whereby NO kills cardiac myocytes is not known. To determine whether NO may trigger programmed cell death (apoptosis) of adult rat ventricular myocytes in culture, the NO donor S-nitroso-N-acetylpenicillamine (SNAP) was shown to kill purified cardiac myocytes in a dose-dependent fashion. In situ analysis of ventricular myocytes plated on chamber slides using nick-end labeling of DNA demonstrated that SNAP induces cardiac myocyte apoptosis, which was confirmed by the identification of oligonucleosomal DNA fragmentation on agarose gel electrophoresis. Similarly, treatment of cardiac myocytes with cytokines that induce inducible NO synthase was shown to cause an NO-dependent induction of apoptosis. Addition of reduced hemoglobin to scavenge NO liberated by SNAP extinguished both the increase in percentage of apoptotic cells and the appearance of DNA ladders. Treatment with SNAP (but not with N-acetylpenicillamine or SNAP + hemoglobin) not only induced apoptosis but resulted in a marked increase in p53 expression in cardiac myocytes detected by Western blotting and immunohistochemistry. These data indicate that NO has the capacity to kill cardiac myocytes by triggering apoptosis and suggest the involvement of p53 in this process.


Subject(s)
Apoptosis/physiology , Heart Ventricles/pathology , Nitric Oxide/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Male , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Rats , Rats, Wistar , Ventricular Function
9.
Am J Cardiol ; 83(8): 1271-4, A9, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10215298

ABSTRACT

Positron emission tomography imaging detected evidence of viability in 17 of 33 patients with evidence of infarction only on their thallium scans who were referred for transplant evaluation. Eleven of these 17 underwent revascularization with excellent outcome.


Subject(s)
Heart Transplantation , Tomography, Emission-Computed , Ventricular Dysfunction, Left/diagnostic imaging , Coronary Angiography , Coronary Artery Bypass , Coronary Disease/complications , Coronary Disease/diagnostic imaging , Coronary Disease/surgery , Follow-Up Studies , Humans , Middle Aged , Observer Variation , Predictive Value of Tests , Retrospective Studies , Severity of Illness Index , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/surgery
10.
J Cardiovasc Pharmacol ; 32(1): 146-55, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9676735

ABSTRACT

Cytokines and cytotoxic agents, including nitric oxide (NO) released by macrophages, play important roles during cardiac allograft rejection. In contrast to agents that suppress T-lymphocyte function, CNI-1493 is a multivalent guanylhydrazone compound that inhibits the synthesis and release of proinflammatory cytokines and NO from macrophages. This study investigated the effects of CNI-1493 on rejecting rat cardiac allografts by using Lewis to Wistar-Furth heterotopic cardiac transplants. CNI-1493 (2 mg/kg i.p., b.i.d.) or vehicle (water) was administered beginning the day before surgery. Rat cardiac allograft survival to cessation of heart beat, apoptosis of cardiac myocytes, degree of myocardial inflammation, and inducible nitric oxide synthase (iNOS) messenger RNA (mRNA), protein, and enzyme activity were studied at days 1, 3, 5, and 7 after transplantation. Allograft survival was increased significantly by 26% from 7.5 +/- 0.8 days in vehicle-treated rats (n = 6) to 9.5 +/- 1.2 days in CNI-1493-treated rats (n = 8, p < 0.05). Apoptotic cells per mm2 myocardium decreased from 2.25 +/- 1.25 to 0.84 +/- 0.49 at day 3 and 31.2 +/- 2.9 to 17.6 +/- 5.43 at day 5 after transplantation with CNI-1493 treatment (p < 0.05). The number of apoptotic myocytes and loss of cardiac muscle cells also decreased significantly at day 5 in the treated animals (p < 0.05). The reduction of myocyte loss at day 5 coincided with a significant decrease of the inflammatory response and reduced macrophage influx (p < 0.05). Myocardial iNOS mRNA, protein, and enzyme levels increased during the course of allograft rejection, and CNI-1493 did not significantly reduce iNOS expression in the rejecting rat allograft. CNI-1493 prolongs allograft survival and reduces myocyte loss, apoptosis, and inflammation during rat cardiac allograft rejection. These effects of CNI-1493 appear to be unrelated to altered NO synthesis but may be related to effects of the drug to inhibit macrophage synthesis of cytokines.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Graft Rejection/drug therapy , Graft Rejection/pathology , Heart Transplantation/physiology , Hydrazones/pharmacology , Myocarditis/drug therapy , Myocarditis/pathology , Myocardium/pathology , Animals , Enzyme Induction/drug effects , Graft Rejection/complications , Male , Myocarditis/etiology , Myocardium/enzymology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Proteins/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Lew , Rats, Inbred WF
11.
Circulation ; 97(23): 2338-45, 1998 Jun 16.
Article in English | MEDLINE | ID: mdl-9639378

ABSTRACT

BACKGROUND: The inducible isoform of the nitric oxide synthase (iNOS) produces large amounts of nitric oxide in response to cytokine stimulation. Previous investigations have demonstrated iNOS expression in the setting of acute and chronic rejection in experimental cardiac transplant models. The goal of this study was to investigate whether iNOS is upregulated in human transplant coronary artery disease (TCAD), a major cause of late mortality after cardiac transplantation. METHODS AND RESULTS: We studied 15 patients with TCAD and 10 with normal coronary arteries. In situ hybridization and immunohistochemistry were used in tissue sections to localize iNOS mRNA and protein, respectively. The presence of peroxynitrite was indirectly assessed by immunostaining with an anti-nitrotyrosine antibody. Normal coronary arteries had no evidence of iNOS expression. In contrast, 30 of 36 coronary artery segments with TCAD (83%) were immunostained by the iNOS antibody. The presence of iNOS mRNA was demonstrated in these vessels by in situ hybridization. Specific cell markers identified iNOS-positive cells as neointimal macrophages and smooth muscle cells. Nitrotyrosine immunoreactivity colocalized with iNOS expression in arteries with TCAD, distributed in macrophages and smooth muscle cells. CONCLUSIONS: iNOS mRNA and protein are expressed in human arteries with TCAD, where they are associated with extensive nitration of protein tyrosines. These findings indicate that the high-output nitric oxide pathway and possibly the oxidant peroxynitrite might be involved in the process leading to the development of TCAD.


Subject(s)
Coronary Disease/metabolism , Heart Transplantation , Macrophages/enzymology , Muscle, Smooth, Vascular/enzymology , Nitric Oxide Synthase/genetics , Adult , Child , Child, Preschool , Coronary Artery Disease/metabolism , Coronary Artery Disease/surgery , Coronary Disease/surgery , Coronary Vessels/chemistry , Coronary Vessels/enzymology , Endothelium, Vascular/chemistry , Endothelium, Vascular/enzymology , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic/physiology , Humans , In Situ Hybridization , Macrophages/chemistry , Male , Middle Aged , Muscle, Smooth, Vascular/chemistry , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/analysis , Tyrosine/analogs & derivatives , Tyrosine/analysis
12.
Transplantation ; 65(6): 804-12, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9539092

ABSTRACT

BACKGROUND: The mechanisms of myocyte death during cardiac allograft rejection are incompletely understood. In a previous study using a rat heterotopic cardiac allograft model, we showed that cardiac myocyte apoptosis, inducible nitric oxide synthase (iNOS) mRNA, protein and enzyme activity, and nitrotyrosine increased simultaneously during cardiac allograft rejection. This study was designed to investigate whether apoptosis and expression of iNOS occur in human cardiac allograft rejection. METHODS: Right ventricular endomyocardial biopsies from 30 cases of allograft rejection (International Society of Heart and Lung Transplantation grade 3A/B) were compared with 12 biopsies with no rejection (International Society of Heart and Lung Transplantation grade 0). Samples were co-labeled for apoptosis and muscle actin. Serial sections were stained for iNOS, nitrotyrosine, and the leukocyte markers CD3, CD4, CD8, and CD68 to identify T-cell subpopulations and macrophages. RESULTS: Biopsies with cardiac allograft rejection showed a 30-fold increase of apoptotic cells when compared with controls. Most apoptotic cardiac myocytes were found in proximity to macrophage (CD68+)-rich inflammatory infiltrates. iNOS immunoreactivity was strongest in macrophages and adjacent myocytes, which also showed high levels of nitrotyrosine, representing damage by peroxynitrite. CONCLUSIONS: Apoptosis is a major form of myocyte death during human cardiac allograft rejection. Cardiac myocyte apoptosis is closely associated with expression of iNOS in macrophages and myocytes and with nitration of myocyte proteins by peroxynitrite.


Subject(s)
Graft Rejection , Heart Transplantation/pathology , Nitric Oxide Synthase/metabolism , Acute Disease , Apoptosis , Biopsy , Humans , Inflammation/pathology , Myocardium/enzymology , Myocardium/pathology , Nitric Oxide Synthase Type II
13.
Arterioscler Thromb Vasc Biol ; 17(10): 1846-58, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9351345

ABSTRACT

The different cell types comprising cardiac muscle express one or more of the three isoforms (neuronal NOS, or nNOS; inducible NOS, or iNOS; and endothelial NOS, or eNOS) of nitric oxide synthase (NOS). nNOS is expressed in orthosympathetic nerve terminals and regulates the release of catecholamines in the heart. eNOS constitutively expressed in endothelial cells inhibits contractile tone and the proliferation of underlying vascular smooth muscle cells, inhibits platelet aggregation and monocyte adhesion, promotes diastolic relaxation, and decreases O2 consumption in cardiac muscle through paracrinally produced NO. eNOS is also constitutively expressed in cardiac myocytes from rodent and human species, where it autocrinally opposes the inotropic action of catecholamines after muscarinic cholinergic and beta-adrenergic receptor stimulation. iNOS gene transcription and protein expression are induced in all cell types after exposure to a variety of inflammatory cytokines. Aside from participating in the immune defense against intracellular microorganisms and viruses, the large amounts of NO produced autocrinally or paracrinally mediate the vasoplegia and myocardial depression characteristic of systemic immune stimulation and promote cell death through apoptosis. In cardiac myocytes, NO may regulate L-type calcium current and contraction through activation of cGMP-dependent protein kinase and cGMP-modulated phosphodiesterases. Other mechanisms independent of cGMP elevations may operate through interaction of NO with heme proteins, non-heme iron, or free thiol residues on target signaling proteins, enzymes, or ion channels. Given the multiplicity of NOS isoforms expressed in cardiac muscle and of the potential molecular targets for the NO produced, tight molecular regulation of NOS expression and activity at the transcriptional and posttranscriptional level appear to be needed to coordinate the many roles of NO in heart function in health and disease.


Subject(s)
Myocardium/enzymology , Nitric Oxide Synthase/physiology , Animals , Endothelium, Vascular/enzymology , Heart/physiology , Humans , Nitric Oxide/physiology
16.
Am J Cardiol ; 80(2): 145-9, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9230149

ABSTRACT

Hemostatic factors are involved in the pathogenesis of native coronary artery disease. However, their role in transplant coronary artery disease is less established. To assess the role of hemostatic factors in transplant coronary artery disease we studied 52 consecutive cardiac transplant patients. The presence of transplant coronary artery disease was determined by angiography. Plasma levels of tissue plasminogen activator (t-PA), plasminogen activator inhibitor-1 (PAI-1), von Willebrand Factor (vWF), and fibrin D-dimer were determined by enzyme-linked immunosorbent assays. Serum lipids were measured by enzymatic methods. Patients with transplant coronary artery disease had higher circulating t-PA (8.6 +/- 0.8 vs. 5.4 +/- 0.6 ng/ml, p = 0.021) and PAI-1 antigen concentrations (38.0 +/- 3.4 vs 25.8 +/- 2.2 ng/ml, p = 0.037). t-PA and PAI-1 antigen concentrations correlated with the severity of angiographic disease (R = 0.34; p = 0.014 for t-PA, and R = 0.45; p = 0.001 for PAI-1). Serum cholesterol levels were higher in patients with transplant coronary artery disease (221 +/- 7.6 vs 191 +/- 9.2 mg/dl, p = 0.039). Serum triglycerides were also higher in patients with transplant coronary artery disease by angiography (246 +/- 38.3 vs 139 +/- 20.8 mg/dl, p = 0.050). Multivariate analysis identified t-PA antigen (p = 0.003) and triglyceride levels (p = 0.038) as independent predictors for the presence of transplant coronary artery disease. We conclude that cardiac transplant patients with evidence of transplant coronary artery disease on coronary angiography have altered hemostatic function which is reflected by elevated levels of circulating t-PA and PAI-1 antigens. The interaction of the hemostatic system and serum lipids in the development of transplant coronary artery disease warrants further study.


Subject(s)
Coronary Disease/blood , Heart Transplantation/physiology , Plasminogen Activator Inhibitor 1/blood , Tissue Plasminogen Activator/blood , Cholesterol/blood , Female , Fibrin Fibrinogen Degradation Products/analysis , Humans , Male , Triglycerides/blood , von Willebrand Factor/analysis
17.
Am J Physiol ; 272(2 Pt 1): C392-8, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9124280

ABSTRACT

Smooth muscle cell (SMC) fibrinolysis is necessary for SMC migration. To determine whether the T cell lymphokines interleukin 4 (IL-4) and interferon-gamma (IFN-gamma) modulate SMC fibrinolysis and migration induced by basic fibroblast growth factor (bFGF), we examined the effects of IL-4 and IFN-gamma on human SMC tissue-type plasminogen activator (tPA), urokinase-type plasminogen activator (UPA), and plasminogen activator inhibitor 1 (PAI-1) antigen production, determined by enzyme-linked immunosorbent assays. Although IL-4 had no effects on SMC tPA, UPA, and PAI-1 production, it potentiated bFGF-induced tPA, UPA, and PAI-1 antigens. IL-4 plus bFGF resulted in a net increase in SMC fibrinolytic activity. IFN-gamma did not significantly affect bFGF induction of SMC tPA and PAI-1 antigens. However, IFN-gamma significantly decreased bFGF-mediated induction of SMC UPA antigen. IFN-gamma decreased the IL-4 plus bFGF induction of both tPA and UPA antigens. IL-4 increased and IFN-gamma abrogated bFGF induction of in vitro SMC migration through a modified micro-Boyden chamber. Therefore, IL-4 and IFN-gamma modulate bFGF-mediated induction of in vitro vascular SMC fibrinolysis and migration.


Subject(s)
Fibrinolysis/physiology , Fibroblast Growth Factor 2/pharmacology , Lymphokines/physiology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , T-Lymphocytes/metabolism , Antigens/analysis , Cell Movement/physiology , Humans , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Muscle, Smooth, Vascular/cytology , Plasminogen Activator Inhibitor 1/immunology , Recombinant Proteins , Tissue Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/immunology
18.
Circulation ; 95(2): 430-7, 1997 Jan 21.
Article in English | MEDLINE | ID: mdl-9008461

ABSTRACT

BACKGROUND: The potential antiatherosclerotic actions of NO were investigated in four groups of mice (n = 10 per group) lacking functional LDL receptor genes, an animal model of familial hypercholesterolemia. Group 1 was fed a regular chow diet. Groups 2 through 4 were fed a 1.25% high-cholesterol diet. In addition, group 3 received supplemental L-arginine and group 4 received L-arginine and N omega-nitro-L-arginine (L-NA), an inhibitor of NO synthase (NOS). METHODS AND RESULTS: Animals were killed at 6 months; aortas were stained with oil red O for planimetry and with antibodies against constitutive and inducible NOSs. Plasma cholesterol was markedly increased in the animals receiving the high-cholesterol diet. Xanthomas appeared in all mice fed the high-cholesterol diet alone but not in those receiving L-arginine. Aortic atherosclerosis was present in all mice on the high-cholesterol diet. The mean atherosclerotic lesion area was reduced significantly (P < .01) in the cholesterol-fed mice given L-arginine compared with those receiving the high-cholesterol diet alone. The mean atherosclerotic lesion area was significantly larger (P < .01) in cholesterol-fed mice receiving L-arginine + L-NA than in those on the high-cholesterol diet alone. Within the atherosclerotic plaques, endothelial cells immunoreacted for endothelial cell NOS; macrophages, foam cells, and smooth muscle cells immunostained strongly for inducible NOS and nitrotyrosine residues. CONCLUSIONS: The data indicate that L-arginine prevents xanthoma formation and reduces atherosclerosis in LDL receptor knockout mice fed a high-cholesterol diet. The abrogation of the beneficial effects of L-arginine by L-NA suggests that the antiatherosclerotic actions of L-arginine are mediated by NOS. The data suggest that L-arginine may be beneficial in familial hypercholesterolemia.


Subject(s)
Arginine/pharmacology , Arteriosclerosis/prevention & control , Mice, Knockout/genetics , Mice, Knockout/physiology , Receptors, LDL/genetics , Xanthomatosis/prevention & control , Animals , Aorta/pathology , Arteriosclerosis/pathology , Cholesterol, Dietary/pharmacology , Female , Hypercholesterolemia/genetics , Male , Mice
19.
Antisense Nucleic Acid Drug Dev ; 7(6): 559-66, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9450913

ABSTRACT

Phosphorothioate oligodeoxynucleotides (PS oligos) manifest both antisense and G-quartet aptameric inhibitory effects on vascular smooth muscle cell (SMC) proliferation. In this study, we examined the effects of three cytidine (S-dC) homopolymers lacking any guanosines of various chain length-S-dC28, S-dC18 and S-dC12-on in vitro SMC proliferation and in vivo neointimal formation. S-dC18 significantly inhibited human vascular SMC proliferation, although it had only half the potency as the same dose of S-dC28. Furthermore, S-dC12 at the same concentrations as S-dC18 did not significantly inhibit vascular SMC proliferation. S-dC28 and S-dC18 inhibited PDGF-induced in vitro SMC migration, whereas D-dC12 had no significant effect on PDGF-induced in vitro SMC migration. We determined the effects of S-dC28, S-dC18, and S-dC12 on neointimal SMC formation in the rat carotid balloon injury model. Rat carotid artery neointimal formation after balloon injury was significantly attenuated by S-dC28 treatment compared with the control group and by S-dC18 treatment compared with the control group. S-dC28 and S-dC18 treatment significantly reduced the intima/media area ratio compared with the values of the control groups. However, S-dC12 did not significantly inhibit neointimal formation. We investigated the time course of the inhibitory effects of S-dC28 on rat carotid artery neointimal formation. S-dC28 significantly inhibited rat carotid artery intimal area and intima/media area ratio at 4 weeks and 8 weeks. Fluoresceinated S-dC28 (FITC-S-dC28) was found to be present throughout the rat carotid arterial wall within 6 hours after balloon injury. Taken together, the potent non-G-quartet, nonsequence-specific inhibitory effects of S-dC compounds on in vitro SMC proliferation and in vivo neointimal formation in the rat carotid balloon injury model are chain length dependent and long lasting.


Subject(s)
Cell Division/drug effects , Cytidine/pharmacology , Endothelium, Vascular/drug effects , Angioplasty, Balloon , Animals , Biopolymers , Carotid Arteries/cytology , Carotid Arteries/metabolism , Cell Movement/drug effects , Cells, Cultured , Cytidine/chemistry , Endothelium, Vascular/cytology , Fluorescein-5-isothiocyanate , Humans , Rats
20.
Circulation ; 94(9): 2096-102, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8901657

ABSTRACT

BACKGROUND: The pathogenesis of transplant coronary artery disease (TCAD) is unknown, but it is thought to derive from an interaction between immune and nonimmune factors, leading to smooth muscle cell proliferation and accumulation in the expanded neointima. Endothelin-1 (ET-1), a potent vasoconstrictor with mitogenic properties for vascular smooth muscle cells, has recently been demonstrated in native vessel atherosclerosis. The present study used immunohistochemistry to investigate the role of ET-1 in TCAD. METHODS AND RESULTS: ET-1 immunoreactivity and cellular localization were assessed in human coronary arteries with TCAD (n = 13) and in normal coronary arteries (n = 10) with single- and double-label immunohistochemistry. The intensity of immunostaining was determined by a semiquantitative method. Diffuse and intense ET-1 immunoreactivity was found in 11 of 13 patients with TCAD (85%), mainly in myointimal cells and, in lesser amounts, in macrophages and endothelial cells. In contrast, normal coronary arteries had only faint immunostaining localized to the endothelial layer. Mean semiquantitative grade was significantly higher in TCAD than in normal arteries (1.8 versus 0.7; P < .05). ET-1 was more frequently present in lipid-rich, atheromatous lesions than in lipid-poor, proliferative ones. Intimal neovessels consistently immunostained for ET-1. CONCLUSIONS: Immunoreactivity for ET-1 is significantly increased in TCAD, possibly as a result of stimulatory cytokines and growth factors that are upregulated in the posttransplant state. The results suggest a role for this mitogenic peptide in the pathogenesis of graft arteriosclerosis.


Subject(s)
Coronary Artery Disease/metabolism , Coronary Disease/metabolism , Endothelin-1/immunology , Heart Transplantation , Adult , Coronary Artery Disease/pathology , Coronary Artery Disease/surgery , Coronary Disease/pathology , Coronary Disease/surgery , Coronary Vessels/chemistry , Coronary Vessels/metabolism , Coronary Vessels/pathology , Endothelin-1/analysis , Endothelin-1/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...